US8613951B2 - Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same - Google Patents

Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same Download PDF

Info

Publication number
US8613951B2
US8613951B2 US12/485,462 US48546209A US8613951B2 US 8613951 B2 US8613951 B2 US 8613951B2 US 48546209 A US48546209 A US 48546209A US 8613951 B2 US8613951 B2 US 8613951B2
Authority
US
United States
Prior art keywords
poly
acid
polymer
therapeutic nanoparticle
nanoparticle
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US12/485,462
Other versions
US20100069426A1 (en
Inventor
Stephen E. Zale
Greg Troiano
Mir Mukkaram Ali
Jeff Hrkach
James Wright
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Bind Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bind Therapeutics Inc filed Critical Bind Therapeutics Inc
Priority to US12/485,462 priority Critical patent/US8613951B2/en
Assigned to BIND BIOSCIENCES, INC. reassignment BIND BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WRIGHT, JAMES, ALI, MIR MUKKARAM, HRKACH, JEFF, TROIANO, GREG, ZALE, STEPHEN E.
Publication of US20100069426A1 publication Critical patent/US20100069426A1/en
Priority to US13/910,328 priority patent/US8623417B1/en
Assigned to BIND THERAPEUTICS, INC. reassignment BIND THERAPEUTICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BIND BIOSCIENCES, INC.
Priority to US14/091,343 priority patent/US9579284B2/en
Application granted granted Critical
Publication of US8613951B2 publication Critical patent/US8613951B2/en
Assigned to PFIZER INC. reassignment PFIZER INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BIND THERAPEUTICS, INC.
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/01Saturated compounds having only one carboxyl group and containing hydroxy or O-metal groups
    • C07C59/08Lactic acid
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/773Nanoparticle, i.e. structure having three dimensions of 100 nm or less

Definitions

  • therapeutics that include an active drug and that are capable of locating in a particular tissue or cell type e.g., a specific diseased tissue, may reduce the amount of the drug in tissues of the body that do not require treatment. This is particularly important when treating a condition such as cancer where it is desirable that a cytotoxic dose of the drug is delivered to cancer cells without killing the surrounding non-cancerous tissue. Further, such therapeutics may reduce the undesirable and sometimes life threatening side effects common in anticancer therapy. For example, nanoparticle therapeutics may, due the small size, evade recognition within the body allowing for targeted and controlled delivery while e.g., remaining stable for an effective amount of time.
  • Therapeutics that offer such therapy and/or controlled release and/or targeted therapy also must be able to deliver an effective amount of drug. It can be a challenge to prepare nanoparticle systems that have an appropriate amount of drug associated each nanoparticle, while keeping the size of the nanoparticles small enough to have advantageous delivery properties. For example, while it is desirable to load a nanoparticle with a high quantity of therapeutic agent, nanoparticle preparations that use a drug load that is too high will result in nanoparticles that are too large for practical therapeutic use. Further, it may be desirable for therapeutic nanoparticles to remain stable so as to e.g. substantially limit rapid or immediate release of the therapeutic agent.
  • the invention provides therapeutic nanoparticle that includes an active agent or therapeutic agent, e.g. an mTOR inhibitor or pharmaceutically acceptable salts thereof, and one, two, or three biocompatible polymers.
  • an active agent or therapeutic agent e.g. an mTOR inhibitor or pharmaceutically acceptable salts thereof
  • one, two, or three biocompatible polymers e.g. an mTOR inhibitor or pharmaceutically acceptable salts thereof.
  • a therapeutic nanoparticle comprising about 1 to about 20 weight percent of a therapeutic agent (such as for example sirolimus, temsirolimus, or everolimus) and about 50 to about 99 weight percent of a biocompatible polymer, e.g. about 70 to about 99 weight percent of a biocompatible polymer.
  • the biocompatible polymer may be a diblock poly(lactic) acid-poly(ethylene)glycol copolymer (e.g.
  • PLA-PEG or a diblock (poly(lactic)-co-poly (glycolic) acid)-poly(ethylene)glycol copolymer (e.g. PLGA-PEG), or the biocompatible polymer may two or more biocompatible polymers, for example, the therapeutic nanoparticles can also include a homopolymer such as poly(lactic) acid homopolymer.
  • a disclosed therapeutic nanoparticle may include about 1 to about 20 weight percent, e.g about 2 to about 20 weight percent or about 10 to about 20 weight percent, of a mTOR inhibitor; and about 70 to about 99 weight percent biocompatible polymer, wherein the biocompatible polymer is selected from the group consisting of a) a diblock poly(lactic) acid-poly(ethylene)glycol copolymer, b) a diblock poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer, c) a combination of a) or b) and a poly (lactic) acid homopolymer or poly(lactic)-co-(glycolic) acid; and d) a combination of a) or b) and a poly (lactic) acid homopolymer or poly(lactic)-co-(glycolic) acid.
  • the diameter of disclosed nanoparticles may be, for example, about 60 to about 120 nm, or about 70 to about 120 nm.
  • Disclosed therapeutic nanoparticles may be stable for at least 5 days at 25° C., e.g. may remain stable over 5 days in vitro, e.g. in a sucrose solution.
  • disclosed particles may substantially immediately release less than about 2% or less than about 5%, or even less than about 10% of the therapeutic agent when placed in a phosphate buffer solution at room temperature, or at 37° C.
  • disclosed nanoparticles may substantially retain the therapeutic agent for at least 5 days at 25° C.
  • disclosed nanoparticles may release the therapeutic agent over a period of at least 1 day or more when administered to a patient.
  • FIG. 1 is flow chart for an emulsion process for forming disclosed nanoparticle.
  • FIG. 2 is a flow diagram for a disclosed emulsion process.
  • FIG. 3 depicts the effect of coarse emulsion preparation on quenched particle size. Placebo organic at 30% solids was used, emulsified at 5:1 W:O using standard aqueous phase (1% sodium cholate, 2% benzyl alcohol, 4% ethyl acetate).
  • FIG. 4 depicts the effect of feed pressure on resultant particle size.
  • FIG. 5 depicts the particle size dependence on scale.
  • Placebo organic phase consisted of 25.5% polymer stock of 50:50 16.5/5 PLA/PEG:8.2 PLA.
  • Organic phase was emulsified 5:1 O:W with standard aqueous phase, and multiple discreet passes were performed, quenching a small portion of emulsion after each pass.
  • the indicated scale represents the total solids of the formulation.
  • FIG. 6 depicts the effect of solids concentration on particle size.
  • FIG. 7 depicts the effect of solids concentration and poly(lactic) homopolymer on loading percentage of sirolimus (rapamycin).
  • FIG. 8 depicts in vitro release of sirolimus over time for disclosed nanoparticles
  • FIG. 9 depicts the effects of poly(lactic) homopolymer on loading percentage of temsirolimus.
  • FIG. 10 depicts the effect of solids concentration on particle size of temsirolimus containing particles.
  • FIG. 11 depicts in vitro release of temsirolimus over time for disclosed nanoparticles
  • the present invention generally relates to polymeric nanoparticles that include an active or therapeutic agent or drug, and methods of making and using such therapeutic nanoparticles.
  • a “nanoparticle” refers to any particle having a diameter of less than 1000 nm, e.g. about 10 nm to about 200 nm.
  • Disclosed therapeutic nanoparticles may include nanoparticles having a diameter of about 60 to about 120 nm, or about 70 to about 130 nm, or about 60 to about 140 nm, or about 70 nm to about 140 nm.
  • Disclosed nanoparticles may include about 0.2 to about 35 weight percent, about 3 to about 40 weight percent, about 5 to about 30 weight percent, about 1 to about 20 weight percent, about 10 to about 30 weight percent, about 5 to about 15 percent, about 15 to 25 weight percent, or even about 4 to about 25 weight percent, e.g. about 10 weight percent of an active agent, such as antineoplastic agent, e.g. a mTOR inhibiting agent (for example sirolimus, temsirolimus or everolimus).
  • an active agent such as antineoplastic agent, e.g. a mTOR inhibiting agent (for example sirolimus, temsirolimus or everolimus).
  • Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers.
  • a contemplated nanoparticle may include about 60 to about 99 weight percent of one, two, three or more biocompatible polymers such as one or more co-polymers (e.g. a diblock polymer) that include a biodegradable polymer (for example poly(lactic)acid and polyethylene glycol, and optionally about 0 to about 50 weight percent of a homopolymer, e.g. biodegradable polymer such as poly(lactic) acid.
  • a biocompatible polymers such as one or more co-polymers (e.g. a diblock polymer) that include a biodegradable polymer (for example poly(lactic)acid and polyethylene glycol, and optionally about 0 to about 50 weight percent of a homopolymer, e.g. biodegradable polymer such as poly(lactic) acid.
  • biodegradable polymer for example poly(lactic)acid and poly
  • disclosed nanoparticles include a matrix of polymers.
  • Disclosed nanoparticles may include one or more polymers, e.g. a diblock co-polymer and/or a monopolymer.
  • Disclosed therapeutic nanoparticles include a therapeutic agent can may associated with the surface of, encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix.
  • the disclosure is directed toward nanoparticles with at least one polymer, for example, a first polymer that may be a co-polymer, e.g. a diblock co-polymer, and optionally a polymer that may be for example a homopolymer.
  • a first polymer that may be a co-polymer, e.g. a diblock co-polymer, and optionally a polymer that may be for example a homopolymer.
  • Polymers can be natural or unnatural (synthetic) polymers.
  • Polymers can be homopolymers or copolymers comprising two or more monomers. In terms of sequence, copolymers can be random, block, or comprise a combination of random and block sequences.
  • Contemplated polymers may be biocompatible and/or biodegradable.
  • polymer is given its ordinary meaning as used in the art, i.e., a molecular structure comprising one or more repeat units (monomers), connected by covalent bonds.
  • the repeat units may all be identical, or in some cases, there may be more than one type of repeat unit present within the polymer.
  • the polymer can be biologically derived, i.e., a biopolymer. Non-limiting examples include peptides or proteins.
  • additional moieties may also be present in the polymer, for example biological moieties such as those described below.
  • the polymer is said to be a “copolymer.” It is to be understood that in any embodiment employing a polymer, the polymer being employed may be a copolymer in some cases.
  • the repeat units forming the copolymer may be arranged in any fashion. For example, the repeat units may be arranged in a random order, in an alternating order, or as a block copolymer, i.e., comprising one or more regions each comprising a first repeat unit (e.g., a first block), and one or more regions each comprising a second repeat unit (e.g., a second block), etc.
  • Block copolymers may have two (a diblock copolymer), three (a triblock copolymer), or more numbers of distinct blocks.
  • Disclosed particles can include copolymers, which, in some embodiments, describes two or more polymers (such as those described herein) that have been associated with each other, usually by covalent bonding of the two or more polymers together.
  • a copolymer may comprise a first polymer and a second polymer, which have been conjugated together to form a block copolymer where the first polymer can be a first block of the block copolymer and the second polymer can be a second block of the block copolymer.
  • a block copolymer may, in some cases, contain multiple blocks of polymer, and that a “block copolymer,” as used herein, is not limited to only block copolymers having only a single first block and a single second block.
  • a block copolymer may comprise a first block comprising a first polymer, a second block comprising a second polymer, and a third block comprising a third polymer or the first polymer, etc.
  • block copolymers can contain any number of first blocks of a first polymer and second blocks of a second polymer (and in certain cases, third blocks, fourth blocks, etc.).
  • block copolymers can also be formed, in some instances, from other block copolymers.
  • a first block copolymer may be conjugated to another polymer (which may be a homopolymer, a biopolymer, another block copolymer, etc.), to form a new block copolymer containing multiple types of blocks, and/or to other moieties (e.g., to non-polymeric moieties).
  • the polymer e.g., copolymer, e.g., block copolymer
  • the polymer can be amphiphilic, i.e., having a hydrophilic portion and a hydrophobic portion, or a relatively hydrophilic portion and a relatively hydrophobic portion.
  • a hydrophilic polymer can be one generally that attracts water and a hydrophobic polymer can be one that generally repels water.
  • a hydrophilic or a hydrophobic polymer can be identified, for example, by preparing a sample of the polymer and measuring its contact angle with water (typically, the polymer will have a contact angle of less than 60°, while a hydrophobic polymer will have a contact angle of greater than about 60°).
  • the hydrophilicity of two or more polymers may be measured relative to each other, i.e., a first polymer may be more hydrophilic than a second polymer.
  • the first polymer may have a smaller contact angle than the second polymer.
  • a polymer e.g., copolymer, e.g., block copolymer
  • a biocompatible polymer i.e., the polymer that does not typically induce an adverse response when inserted or injected into a living subject, for example, without significant inflammation and/or acute rejection of the polymer by the immune system, for instance, via a T-cell response.
  • the therapeutic particles contemplated herein can be non-immunogenic.
  • non-immunogenic refers to endogenous growth factor in its native state which normally elicits no, or only minimal levels of, circulating antibodies, T-cells, or reactive immune cells, and which normally does not elicit in the individual an immune response against itself.
  • Biocompatibility typically refers to the acute rejection of material by at least a portion of the immune system, i.e., a nonbiocompatible material implanted into a subject provokes an immune response in the subject that can be severe enough such that the rejection of the material by the immune system cannot be adequately controlled, and often is of a degree such that the material must be removed from the subject.
  • One simple test to determine biocompatibility can be to expose a polymer to cells in vitro; biocompatible polymers are polymers that typically will not result in significant cell death at moderate concentrations, e.g., at concentrations of 50 micrograms/10 6 cells.
  • a biocompatible polymer may cause less than about 20% cell death when exposed to cells such as fibroblasts or epithelial cells, even if phagocytosed or otherwise uptaken by such cells.
  • biocompatible polymers include polydioxanone (PDO), polyhydroxyalkanoate, polyhydroxybutyrate, poly(glycerol sebacate), polyglycolide, polylactide, PLGA, polycaprolactone, or copolymers or derivatives including these and/or other polymers.
  • contemplated biocompatible polymers may be biodegradable, i.e., the polymer is able to degrade, chemically and/or biologically, within a physiological environment, such as within the body.
  • biodegradable polymers are those that, when introduced into cells, are broken down by the cellular machinery (biologically degradable) and/or by a chemical process, such as hydrolysis, (chemically degradable) into components that the cells can either reuse or dispose of without significant toxic effect on the cells.
  • the biodegradable polymer and their degradation byproducts can be biocompatible.
  • a contemplated polymer may be one that hydrolyzes spontaneously upon exposure to water (e.g., within a subject), the polymer may degrade upon exposure to heat (e.g., at temperatures of about 37° C.). Degradation of a polymer may occur at varying rates, depending on the polymer or copolymer used. For example, the half-life of the polymer (the time at which 50% of the polymer can be degraded into monomers and/or other non-polymeric moieties) may be on the order of days, weeks, months, or years, depending on the polymer.
  • the polymers may be biologically degraded, e.g., by enzymatic activity or cellular machinery, in some cases, for example, through exposure to a lysozyme (e.g., having relatively low pH).
  • the polymers may be broken down into monomers and/or other non-polymeric moieties that cells can either reuse or dispose of without significant toxic effect on the cells (for example, polylactide may be hydrolyzed to form lactic acid, polyglycolide may be hydrolyzed to form glycolic acid, etc.).
  • polymers may be polyesters, including copolymers comprising lactic acid and glycolic acid units, such as poly(lactic acid-co-glycolic acid) and poly(lactide-co-glycolide), collectively referred to herein as “PLGA”; and homopolymers comprising glycolic acid units, referred to herein as “PGA,” and lactic acid units, such as poly-L-lactic acid, poly-D-lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as “PLA.”
  • exemplary polyesters include, for example, polyhydroxyacids; PEGylated polymers and copolymers of lactide and glycolide (e.g., PEGylated PLA, PEGylated PGA, PEGylated PLGA, and derivatives thereof.
  • polyesters include, for example, polyanhydrides, poly(ortho ester) PEGylated poly(ortho ester), poly(caprolactone), PEGylated poly(caprolactone), polylysine, PEGylated polylysine, poly(ethylene imine), PEGylated poly(ethylene imine), poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[ ⁇ -(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
  • a polymer may be PLGA.
  • PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA can be characterized by the ratio of lactic acid:glycolic acid.
  • Lactic acid can be L-lactic acid, D-lactic acid, or D,L-lactic acid.
  • the degradation rate of PLGA can be adjusted by altering the lactic acid-glycolic acid ratio.
  • PLGA to be used in accordance with the present invention can be characterized by a lactic acid:glycolic acid molar ratio of approximately 85:15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
  • the ratio of lactic acid to glycolic acid monomers in the polymer of the particle may be selected to optimize for various parameters such as water uptake, therapeutic agent release and/or polymer degradation kinetics can be optimized.
  • polymers may be one or more acrylic polymers.
  • acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid polyacrylamide, amino alkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers.
  • the acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.
  • polymers can be cationic polymers.
  • cationic polymers are able to condense and/or protect negatively charged strands of nucleic acids (e.g. DNA, RNA, or derivatives thereof).
  • Amine-containing polymers such as poly(lysine), polyethylene imine (PEI), and poly(amidoamine) dendrimers are contemplated for use, in some embodiments, in a disclosed particle.
  • polymers can be degradable polyesters bearing cationic side chains.
  • polyesters include poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester).
  • a polymer e.g., copolymer, e.g., block copolymer
  • poly(ethylene glycol) repeat units can also be referred to as a “PEGylated” polymer.
  • Such polymers can control inflammation and/or immunogenicity (i.e., the ability to provoke an immune response) and/or lower the rate of clearance from the circulatory system via the reticuloendothelial system (RES), due to the presence of the poly(ethylene glycol) groups.
  • RES reticuloendothelial system
  • PEGylation may also be used, in some cases, to decrease charge interaction between a polymer and a biological moiety, e.g., by creating a hydrophilic layer on the surface of the polymer, which may shield the polymer from interacting with the biological moiety.
  • the addition of poly(ethylene glycol) repeat units may increase plasma half-life of the polymer (e.g., copolymer, e.g., block copolymer), for instance, by decreasing the uptake of the polymer by the phagocytic system while decreasing transfection/uptake efficiency by cells.
  • Particles disclosed herein may or may not contain PEG.
  • certain embodiments can be directed towards copolymers containing poly(ester-ether)s, e.g., polymers having repeat units joined by ester bonds (e.g., R—C(O)—O—R′ bonds) and ether bonds (e.g., R—O—R′ bonds).
  • a biodegradable polymer such as a hydrolyzable polymer, containing carboxylic acid groups, may be conjugated with poly(ethylene glycol) repeat units to form a poly(ester-ether).
  • PEG may include a terminal end group, for example, when PEG is not conjugated to a ligand.
  • PEG may terminate in a hydroxyl, a methoxy or other alkoxyl group, a methyl or other alkyl group, an aryl group, a carboxylic acid, an amine, an amide, an acetyl group, a guanidino group, or an imidazole.
  • Other contemplated end groups include azide, alkyne, maleimide, aldehyde, hydrazide, hydroxylamine, alkoxyamine, or thiol moieties.
  • the molecular weight of the polymers can be optimized for effective treatment as disclosed herein.
  • the molecular weight of a polymer may influence particle degradation rate (such as when the molecular weight of a biodegradable polymer can be adjusted), solubility, water uptake, and drug release kinetics.
  • the molecular weight of the polymer can be adjusted such that the particle biodegrades in the subject being treated within a reasonable period of time (ranging from a few hours to 1-2 weeks, 3-4 weeks, 5-6 weeks, 7-8 weeks, etc.).
  • a disclosed particle can for example comprise a copolymer of PEG and PLGA
  • the PEG can have a molecular weight of 1,000-20,000, e.g., 5,000-20,000, e.g., 10,000-20,000
  • the PLGA can have a molecular weight of 5,000-100,000, e.g., 20,000-70,000, e.g., 20,000-50,000.
  • an exemplary therapeutic nanoparticle that includes about 10 to about 99 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer, or about 20 to about 80 weight percent, about 40 to about 80 weight percent, or about 30 to about 50 weight percent, or about 70 to about 90 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer.
  • Exemplary poly(lactic) acid-poly(ethylene)glycol copolymers can include a number average molecular weight of about 15 to about 20 kDa, or about 10 to about 25 kDa of poly(lactic) acid and a number average molecular weight of about 4 to about 6, or about 2 kDa to about 10 kDa of poly(ethylene)glycol.
  • Disclosed nanoparticles may optionally include about 1 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid (which does not include PEG, e.g a homopolymer of PLA), or may optionally include about 1 to about 50 weight percent, or about 10 to about 50 weight percent or about 30 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid.
  • poly(lactic) or poly(lactic)-co-poly(glycolic) acid may have a number average molecule weight of about 5 to about 15 kDa, or about 5 to about 12 kDa.
  • Exemplary homopolymeric PLA may have a number average molecular weight of about 5 to about 10 kDa.
  • Exemplary PLGA may have a number average molecular weight of about 8 to about 12 kDa.
  • disclosed polymers of may be conjugated to a lipid, e.g. “end-capped,” for example, may include a lipid-terminated PEG.
  • the lipid portion of the polymer can be used for self assembly with another polymer, facilitating the formation of a nanoparticle.
  • a hydrophilic polymer could be conjugated to a lipid that will self assemble with a hydrophobic polymer.
  • Exemplary lipids include fatty acids such as long chain (e.g., C 8 -C 50 ), substituted or unsubstituted hydrocarbons.
  • a fatty acid group can be a C 10 -C 20 fatty acid or salt thereof.
  • a fatty acid group can be a C 15 -C 20 fatty acid or salt thereof.
  • a fatty acid can be unsaturated, monounsaturated, or polyunsaturated.
  • a fatty acid group can be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid.
  • a fatty acid group can be one or more of palmitoleic, oleic, vaccenic, linoleic, alpha-linolenic, gamma-linoleic, arachidonic, gadoleic, arachidonic, eicosapentaenoic, docosahexaenoic, or erucic acid.
  • the lipid is of the Formula V:
  • the lipid is 1,2 distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), and salts thereof, e.g., the sodium salt.
  • DSPE 1,2 distearoyl-sn-glycero-3-phosphoethanolamine
  • optional small molecule targeting moieties are bonded, e.g., covalently bonded, to the lipid component of the nanoparticle.
  • a nanoparticle comprising a therapeutic agent, a polymeric matrix comprising functionalized and non-functionalized polymers, a lipid, and a low-molecular weight targeting ligand, wherein the targeting ligand is bonded, e.g., covalently bonded, to the lipid component of the nanoparticle.
  • nanoparticles may include an optional targeting moiety, i.e., a moiety able to bind to or otherwise associate with a biological entity, for example, a membrane component, a cell surface receptor, prostate specific membrane antigen, or the like.
  • a targeting moiety present on the surface of the particle may allow the particle to become localized at a particular targeting site, for instance, a tumor, a disease site, a tissue, an organ, a type of cell, etc.
  • the drug or other payload may then, in some cases, be released from the particle and allowed to interact locally with the particular targeting site.
  • the targeting moiety may be a low-molecular weight ligand, e.g., a low-molecular weight PSMA ligand.
  • a targeting portion may cause the particles to become localized to a tumor, a disease site, a tissue, an organ, a type of cell, etc. within the body of a subject, depending on the targeting moiety used.
  • a low-molecular weight PSMA ligand may become localized to prostate cancer cells.
  • the subject may be a human or non-human animal.
  • subjects include, but are not limited to, a mammal such as a dog, a cat, a horse, a donkey, a rabbit, a cow, a pig, a sheep, a goat, a rat, a mouse, a guinea pig, a hamster, a primate, a human or the like.
  • a mammal such as a dog, a cat, a horse, a donkey, a rabbit, a cow, a pig, a sheep, a goat, a rat, a mouse, a guinea pig, a hamster, a primate, a human or the like.
  • Contemplated targeting moieties include small molecules.
  • the term “small molecule” refers to organic compounds, whether naturally-occurring or artificially created (e.g., via chemical synthesis) that have relatively low molecular weight and that are not proteins, polypeptides, or nucleic acids. Small molecules typically have multiple carbon-carbon bonds.
  • small molecules are less than about 2000 g/mol in size. In some embodiments, small molecules are less than about 1500 g/mol or less than about 1000 g/mol. In some embodiments, small molecules are less than about 800 g/mol or less than about 500 g/mol, for example about 100 g/mol to about 600 g/mol, or about 200 g/mol to about 500 g/mol.
  • a ligand may be a the low-molecular weight PSMA ligand such as
  • small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include PSMA peptidase inhibitors such as 2-PMPA, GPI5232, VA-033, phenylalkylphosphonamidates and/or analogs and derivatives thereof.
  • small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include thiol and indole thiol derivatives, such as 2-MPPA and 3-(2-mercaptoethyl)-1H-indole-2-carboxylic acid derivatives.
  • small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include hydroxamate derivatives.
  • small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include PBDA- and urea-based inhibitors, such as ZJ 43, ZJ 11, ZJ 17, ZJ 38 and/or and analogs and derivatives thereof, androgen receptor targeting agents (ARTAs), polyamines, such as putrescine, spermine, and spermidine, inhibitors of the enzyme glutamate carboxylase II (GCPII), also known as NAAG Peptidase or NAALADase.
  • PBDA- and urea-based inhibitors such as ZJ 43, ZJ 11, ZJ 17, ZJ 38 and/or and analogs and derivatives thereof
  • ARTAs androgen receptor targeting agents
  • polyamines such as putrescine, spermine, and spermidine
  • GCPII glutamate carboxylase II
  • the targeting moiety can be a ligand that targets Her2, EGFR, or toll receptors.
  • the targeting moieties may include a nucleic acid, polypeptide, glycoprotein, carbohydrate, or lipid.
  • a targeting moiety can be a nucleic acid targeting moiety (e.g. an aptamer, e.g., the A10 aptamer) that binds to a cell type specific marker.
  • an aptamer is an oligonucleotide (e.g., DNA, RNA, or an analog or derivative thereof) that binds to a particular target, such as a polypeptide.
  • a targeting moiety may be a naturally occurring or synthetic ligand for a cell surface receptor, e.g., a growth factor, hormone, LDL, transferrin, etc.
  • a targeting moiety can be an antibody, which term is intended to include antibody fragments, characteristic portions of antibodies, single chain targeting moieties can be identified, e.g., using procedures such as phage display.
  • Targeting moieties may be a targeting peptide or targeting peptidomimetic has a length of up to about 50 residues.
  • a targeting moieties may include the amino acid sequence AKERC, CREKA, ARYLQKLN or AXYLZZLN, wherein X and Z are variable amino acids, or conservative variants or peptidomimetics thereof.
  • the targeting moiety is a peptide that includes the amino acid sequence AKERC, CREKA, ARYLQKLN or AXYLZZLN, wherein X and Z are variable amino acids, and has a length of less than 20, 50 or 100 residues.
  • the CREKA (Cys Arg Glu Lys Ala) peptide or a peptidomimetic thereof peptide or the octapeptide AXYLZZLN are also contemplated as targeting moieties, as well as peptides, or conservative variants or peptidomimetics thereof, that binds or forms a complex with collagen IV, or the targets tissue basement membrane (e.g., the basement membrane of a blood vessel), can be used as a targeting moiety.
  • Exemplary targeting moieties include peptides that target ICAM (intercellular adhesion molecule, e.g. ICAM-1).
  • Targeting moieties disclosed herein are typically conjugated to a disclosed polymer or copolymer (e.g. PLA-PEG), and such a polymer conjugate may form part of a disclosed nanoparticle.
  • a disclosed therapeutic nanoparticle may optionally include about 0.2 to about 10 weight percent of a PLA-PEG or PLGA-PEG, wherein the PEG is functionalized with a targeting ligand.
  • Contemplated therapeutic nanoparticles may include, for example, about 0.2 to about 10 mole percent PLA-PEG-ligand or poly (lactic) acid co poly (glycolic) acid-PEG-ligand.
  • PLA-PEG-ligand may include a PLA with a number average molecular weight of about 10 kDa to about 20 kDa and PEG with a number average molecular weight of about 4,000 to about 8,000.
  • Disclosed nanoparticles may have a substantially spherical (i.e., the particles generally appear to be spherical), or non-spherical configuration.
  • the particles upon swelling or shrinkage, may adopt a non-spherical configuration.
  • the particles may include polymeric blends.
  • a polymer blend may include a first co-polymer that includes polyethylene glycol and a second polymer.
  • Disclosed nanoparticles may have a characteristic dimension of less than about 1 micrometer, where the characteristic dimension of a particle is the diameter of a perfect sphere having the same volume as the particle.
  • the particle can have a characteristic dimension of the particle can be less than about 300 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, less than about 50 nm, less than about 30 nm, less than about 10 nm, less than about 3 nm, or less than about 1 nm in some cases.
  • disclosed nanoparticles may have a diameter of about 70 nm-200 nm, or about 70 nm to about 180 nm, about 80 nm to about 130 nm, about 80 nm to about 120 nm.
  • the particles can have an interior and a surface, where the surface has a composition different from the interior, i.e., there may be at least one compound present in the interior but not present on the surface (or vice versa), and/or at least one compound is present in the interior and on the surface at differing concentrations.
  • a compound such as a targeting moiety (i.e., a low-molecular weight ligand) of a polymeric conjugate of the present invention, may be present in both the interior and the surface of the particle, but at a higher concentration on the surface than in the interior of the particle, although in some cases, the concentration in the interior of the particle may be essentially nonzero, i.e., there is a detectable amount of the compound present in the interior of the particle.
  • a targeting moiety i.e., a low-molecular weight ligand
  • the interior of the particle is more hydrophobic than the surface of the particle.
  • the interior of the particle may be relatively hydrophobic with respect to the surface of the particle, and a drug or other payload may be hydrophobic, and readily associates with the relatively hydrophobic center of the particle.
  • the drug or other payload can thus be contained within the interior of the particle, which can shelter it from the external environment surrounding the particle (or vice versa).
  • a drug or other payload contained within a particle administered to a subject will be protected from a subject's body, and the body may also be substantially isolated from the drug for at least a period of time.
  • a therapeutic polymeric nanoparticle comprising a first non-functionalized polymer; an optional second non-functionalized polymer; an optional functionalized polymer comprising a targeting moiety; and a therapeutic agent
  • the first non-functionalized polymer is PLA, PLGA, or PEG, or copolymers thereof, e.g. a diblock co-polymer PLA-PEG.
  • exemplary nanoparticle may have a PEG corona with a density of about 0.065 g/cm 3 , or about 0.01 to about 0.10 g/cm 3 .
  • Disclosed nanoparticles may be stable (e.g. retain substantially all active agent) for example in a solution that may contain a saccharide, for at least about 3 days, about 4 days or at least about 5 days at room temperature, or at 25° C.
  • disclosed nanoparticles may also include a fatty alcohol, which may increase the rate of drug release.
  • disclosed nanoparticles may include a C 8 -C 30 alcohol such as cetyl alcohol, octanol, stearyl alcohol, arachidyl alcohol, docosonal, or octasonal.
  • Nanoparticles may have controlled release properties, e.g., may be capable of delivering an amount of active agent to a patient, e.g., to specific site in a patient, over an extended period of time, e.g. over 1 day, 1 week, or more.
  • disclosed nanoparticles substantially immediately releases (e.g. over about 1 minute to about 30 minutes) less than about 2%, less than about 4%, less than about 5%, or less than about 10% of an active agent (e.g. a taxane) agent, for example when places in a phosphate buffer solution at room temperature and/or at 37° C.
  • an active agent e.g. a taxane
  • the invention comprises a nanoparticle comprising 1) a polymeric matrix and 2) an amphiphilic compound or layer that surrounds or is dispersed within the polymeric matrix forming a continuous or discontinuous shell for the particle,
  • An amphiphilic layer can reduce water penetration into the nanoparticle, thereby enhancing drug encapsulation efficiency and slowing drug release. Further, these amphipilic layer protected nanoparticles can provide therapeutic advantages by releasing the encapsulated drug and polymer at appropriate times.
  • amphiphilic refers to a property where a molecule has both a polar portion and a non-polar portion. Often, an amphiphilic compound has a polar head attached to a long hydrophobic tail. In some embodiments, the polar portion is soluble in water, while the non-polar portion is insoluble in water. In addition, the polar portion may have either a formal positive charge, or a formal negative charge. Alternatively, the polar portion may have both a formal positive and a negative charge, and be a zwitterion or inner salt.
  • Exemplary amphiphilic compound include, for example, one or a plurality of the following: naturally derived lipids, surfactants, or synthesized compounds with both hydrophilic and hydrophobic moieties.
  • amphiphilic compounds include, but are not limited to, phospholipids, such as 1,2 distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcholine (DBPC), ditricosanoylphosphatidylcholine (DTPC), and dilignoceroylphatidylcholine (DLPC), incorporated at a ratio of between 0.01-60 (weight lipid/w polymer), most preferably between 0.1-30 (weight lipid/w polymer).
  • DSPE dipalmitoylphosphatidylcholine
  • DSPC distearoylphosphatidylcholine
  • DAPC diarachidoylphosphatidylcholine
  • DBPC dibehenoylphosphatid
  • Phospholipids which may be used include, but are not limited to, phosphatidic acids, phosphatidyl cholines with both saturated and unsaturated lipids, phosphatidyl ethanolamines, phosphatidylglycerols, phosphatidylserines, phosphatidylinositols, lysophosphatidyl derivatives, cardiolipin, and ⁇ -acyl-y-alkyl phospholipids.
  • phospholipids include, but are not limited to, phosphatidylcholines such as dioleoylphosphatidylcholine, dimyristoylphosphatidylcholine, dipentadecanoylphosphatidylcholine dilauroylphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcho-line (DBPC), ditricosanoylphosphatidylcholine (DTPC), dilignoceroylphatidylcholine (DLPC); and phosphatidylethanolamines such as dioleoylphosphatidylethanolamine or 1-hexadecyl-2-palmitoylglycerophos-phoethanolamine. Synthetic phospholipids with asymmetric acyl chains (e.
  • an amphiphilic component may include lecithin, and/or in particular, phosphatidylcholine.
  • Another aspect of the invention is directed to systems and methods of making disclosed nanoparticles.
  • using two or more different polymers e.g., a copolymer such as a diblock copolymer and a homopolymer
  • properties of particles may be controlled.
  • the methods described herein form nanoparticles that have a high amount of encapsulated therapeutic agent, for example, may include about 1 to about 40 weight percent, or about 1 to about 30 weight percent, e.g. about 10 to about 25 weight percent or about 5 to about 20 weight percent therapeutic agent.
  • a nanoemulsion process is provided, such as the process represented in FIGS. 1 and 2 .
  • a therapeutic agent for example, PLA-PEG or PLGA-PEG
  • a second polymer e.g. (PL(G)A or PLA
  • an organic solution to form a first organic phase.
  • Such first phase may include about 5 to about 50% weight solids, e.g about 5 to about 40% solids, or about 10 to about 30% solids, e.g. about 10%, 15%, 20% solids.
  • the first organic phase may be combined with a first aqueous solution to form a second phase.
  • the organic solution can include, for example, acetonitrile, tetrahydrofuran, ethyl acetate, isopropyl alcohol, isopropyl acetate, dimethylformamide, methylene chloride, dichloromethane, chloroform, acetone, benzyl alcohol, Tween 80, Span 80, or the like, and combinations thereof.
  • the organic phase may include benzyl alcohol, ethyl acetate, and combinations thereof.
  • the second phase can be between about 1 and 50 weight %, e.g., 5-40 weight %, solids.
  • the aqueous solution can be water, optionally in combination with one or more of sodium cholate, ethyl acetate, and benzyl alcohol.
  • the oil or organic phase may use solvent that is only partially miscible with the nonsolvent (water). Therefore, when mixed at a low enough ratio and/or when using water pre-saturated with the organic solvents, the oil phase remains liquid.
  • the oil phase may bee emulsified into an aqueous solution and, as liquid droplets, sheared into nanoparticles using, for example, high energy dispersion systems, such as homogenizers or sonicators.
  • the aqueous portion of the emulsion, otherwise known as the “water phase” may be surfactant solution consisting of sodium cholate and pre-saturated with ethyl acetate and benzyl alcohol.
  • Emulsifying the second phase to form an emulsion phase may be performed in one or two emulsification steps.
  • a primary emulsion may be prepared, and then emulsified to form a fine emulsion.
  • the primary emulsion can be formed, for example, using simple mixing, a high pressure homogenizer, probe sonicator, stir bar, or a rotor stator homogenizer.
  • the primary emulsion may be formed into a fine emulsion through the use of e.g. probe sonicator or a high pressure homogenizer, e.g. by using 1, 2, 3 or more passes through a homogenizer.
  • the pressure used may be about 4000 to about 8000 psi, or about 4000 to about 5000 psi, e.g. 4000 or 5000 psi.
  • a solvent dilution via aqueous quench may be used.
  • the emulsion can be diluted into cold water to a concentration sufficient to dissolve all of the organic solvent to form a quenched phase.
  • Quenching may be performed at least partially at a temperature of about 5° C. or less.
  • water used in the quenching may be at a temperature that is less that room temperature (e.g. about 0 to about 10° C., or about 0 to about 5° C.).
  • not all of the therapeutic agent is encapsulated in the particles at this stage, and a drug solubilizer is added to the quenched phase to form a solubilized phase.
  • the drug solubilizer may be for example, Tween 80, Tween 20, polyvinyl pyrrolidone, cyclodextran, sodium dodecyl sulfate, or sodium cholate.
  • Tween-80 may added to the quenched nanoparticle suspension to solubilize the free drug and prevent the formation of drug crystals.
  • a ratio of drug solubilizer to therapeutic agent is about 100:1 to about 10:1.
  • the solubilized phase may be filtered to recover the nanoparticles.
  • ultrafiltration membranes may be used to concentrate the nanoparticle suspension and substantially eliminate organic solvent, free drug, and other processing aids (surfactants).
  • Exemplary filtration may be performed using a tangential flow filtration system.
  • a membrane with a pore size suitable to retain nanoparticles while allowing solutes, micelles, and organic solvent to pass nanoparticles can be selectively separated.
  • Exemplary membranes with molecular weight cut-offs of about 300-500 kDa ( ⁇ 5-25 nm) may be used.
  • Diafiltration may be performed using a constant volume approach, meaning the diafiltrate (cold deionized water, e.g. about 0° C. to about 5° C., or 0 to about 10° C.) may added to the feed suspension at the same rate as the filtrate is removed from the suspension.
  • filtering may include a first filtering using a first temperature of about 0 to about 5° C., or 0° C. to about 10° C., and a second temperature of about 20° C. to about 30° C., or 15° C. to about 35° C.
  • filtering may include processing about 1 to about 6 diavolumes at about 0 to about 5° C., and processing at least one diavolume (e.g. about 1 to about 3 or about 1-2 diavolumes) at about 20° C. to about 30° C.
  • the particles may be passed through one, two or more sterilizing and/or depth filters, for example, using ⁇ 0.2 ⁇ m depth pre-filter.
  • an organic phase is formed composed of a mixture of a therapeutic agent, e.g., sirolimus, and polymer (homopolymer, and co-polymer).
  • the organic phase may be mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and optionally dissolved solvent.
  • a primary emulsion may then formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer. The primary emulsion is then formed into a fine emulsion through the use of e.g. high pressure homogenizer.
  • Such fine emulsion may then quenched by, e.g. addition to deionized water under mixing.
  • An exemplary quench:emulsion ratio may be about approximately 8.5:1.
  • a solution of Tween e.g., Tween 80
  • Tween 80 can then be added to the quench to achieve e.g. approximately 2% Tween overall, which may serves to dissolve free, unencapsulated drug.
  • Formed nanoparticles may then be isolated through either centrifugation or ultrafiltration/diafiltration.
  • any agents including, for example, therapeutic agents (e.g. anti-cancer agents), diagnostic agents (e.g. contrast agents; radionuclides; and fluorescent, luminescent, and magnetic moieties), prophylactic agents (e.g. vaccines), and/or nutraceutical agents (e.g. vitamins, minerals, etc.) may be delivered by the disclosed nanoparticles.
  • therapeutic agents e.g. anti-cancer agents
  • diagnostic agents e.g. contrast agents; radionuclides; and fluorescent, luminescent, and magnetic moieties
  • prophylactic agents e.g. vaccines
  • nutraceutical agents e.g. vitamins, minerals, etc.
  • nutraceutical agents e.g. vitamins, minerals, etc.
  • the agent to be delivered is an agent useful in the treatment of cancer (e.g., an anti-neoplastic agent).
  • controlled release is generally meant to encompass release of a substance (e.g., a drug) at a selected site or otherwise controllable in rate, interval, and/or amount.
  • Controlled release encompasses, but is not necessarily limited to, substantially continuous delivery, patterned delivery (e.g., intermittent delivery over a period of time that is interrupted by regular or irregular time intervals), and delivery of a bolus of a selected substance (e.g., as a predetermined, discrete amount if a substance over a relatively short period of time (e.g., a few seconds or minutes)).
  • the active agent or drug may be a therapeutic agent such as an mTOR (mammalian target of rapamycin) inhibitor such as sirolimus (rapamycin), temsirolimus, or everolimus, a taxane or diterpene derivative such as paclitaxel (or its derivatives such as DHA-paclitaxel or PG-paxlitaxel) or docetaxel.
  • mTOR mimmalian target of rapamycin
  • rapamycin sirolimus
  • temsirolimus temsirolimus
  • everolimus a taxane or diterpene derivative
  • paclitaxel or its derivatives such as DHA-paclitaxel or PG-paxlitaxel
  • docetaxel paclitaxel (or its derivatives such as DHA-paclitaxel or PG-paxlitaxel) or docetaxel.
  • the active agent or drug may be a vinca alkaloid such as vinore
  • Nanoparticles disclosed herein may be combined with pharmaceutical acceptable carriers to form a pharmaceutical composition.
  • the carriers may be chosen based on the route of administration as described below, the location of the target issue, the drug being delivered, the time course of delivery of the drug, etc.
  • compositions and particles disclosed herein can be administered to a patient by any means known in the art including oral and parenteral routes.
  • patient refers to humans as well as non-humans, including, for example, mammals, birds, reptiles, amphibians, and fish.
  • the non-humans may be mammals (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a primate, or a pig).
  • parenteral routes are desirable since they avoid contact with the digestive enzymes that are found in the alimentary canal.
  • inventive compositions may be administered by injection (e.g., intravenous, subcutaneous or intramuscular, intraperitoneal injection), rectally, vaginally, topically (as by powders, creams, ointments, or drops), or by inhalation (as by sprays).
  • injection e.g., intravenous, subcutaneous or intramuscular, intraperitoneal injection
  • rectally rectally, vaginally, topically (as by powders, creams, ointments, or drops), or by inhalation (as by sprays).
  • disclosed nanoparticles may be administered to a subject in need thereof systemically, e.g., by IV infusion or injection.
  • sterile injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension, or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the inventive conjugate is suspended in a carrier fluid comprising 1% (w/v) sodium carboxymethyl cellulose and 0.1% (v/v) TWEENTM 80.
  • the injectable formulations can be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the encapsulated or unencapsulated conjugate is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, (c) humectants such as glycerol, (d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, (e) solution retarding agents such as paraffin, (f) absorption accelerators such as quaternary ammonium compounds, (g) wetting agents such as, for example
  • Disclosed nanoparticles may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of nanoparticle appropriate for the patient to be treated.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. An animal model may also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • Therapeutic efficacy and toxicity of nanoparticles can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED 50 (the dose is therapeutically effective in 50% of the population) and LD 50 (the dose is lethal to 50% of the population).
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 50 /ED 50 .
  • Pharmaceutical compositions which exhibit large therapeutic indices may be useful in some embodiments.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for human use.
  • a pharmaceutical composition in an exemplary embodiment, includes a plurality of nanoparticles each comprising a therapeutic agent; and a pharmaceutically acceptable excipient.
  • a composition suitable for freezing is comtemplated, including nanoparticles disclosed herein and a solution suitable for freezing, e.g., a sugar (e.g. sucrose) solution is added to a nanoparticle suspension.
  • a sugar e.g. sucrose
  • the sucrose may e.g., act as a cryoprotectant to prevent the particles from aggregating upon freezing.
  • a nanoparticle formulation comprising a plurality of disclosed nanoparticles, sucrose and water; wherein, for example, the nanoparticles/sucrose/water is are present with about 5-10%/10-15%/80-90% (w/w/w).
  • therapeutic particles disclosed herein may be used to treat, alleviate, ameliorate, relieve, delay onset of, inhibit progression of, reduce severity of, and/or reduce incidence of one or more symptoms or features of a disease, disorder, and/or condition.
  • disclosed therapeutic particles that include e.g., temsirolimus may be used to treat renal cell carcinoma.
  • disclosed therapeutic particles that include e.g. everolimus or temsirolimus may be used to treat kidney cancer, glioblastoma multiforme, mantle cell lymphoma, or dermal Kaposi's sarcoma.
  • Other methods contemplated herein include methods of treating patients having tuberous sclerosis complex, and/or autism by administering an effective amount of a disclosed nanoparticle.
  • Methods contemplated herein include, for example, a method of preventing or deterring neointimal hyperplasia in a blood vessel of a patient, for example, a patient receiving a bare metal stent in a lesion of the blood vessel, is disclosed, comprising administering a composition comprising disclosed therapeutic particles such as those that include sirolimus or everolimus. Also contemplated herein are methods of treating or preventing restenosis (e.g. in a patient receiving a stent) comprising administering disclosed nanoparticles having e.g. sirolimus or everolimus to a patient.
  • Disclosed treatment methods may comprise administering a therapeutically effective amount of the disclosed therapeutic particles to a subject in need thereof, in such amounts and for such time as is necessary to achieve the desired result.
  • a “therapeutically effective amount” is that amount effective for treating, alleviating, ameliorating, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of e.g. a cancer being treated.
  • therapeutic protocols that include administering a therapeutically effective amount of an disclosed therapeutic particle to a healthy individual (i.e., a subject who does not display any symptoms of cancer and/or who has not been diagnosed with cancer).
  • healthy individuals may be “immunized” with an inventive targeted particle prior to development of cancer and/or onset of symptoms of cancer; at risk individuals (e.g., patients who have a family history of cancer; patients carrying one or more genetic mutations associated with development of cancer; patients having a genetic polymorphism associated with development of cancer; patients infected by a virus associated with development of cancer; patients with habits and/or lifestyles associated with development of cancer; etc.) can be treated substantially contemporaneously with (e.g., within 48 hours, within 24 hours, or within 12 hours of) the onset of symptoms of cancer.
  • individuals known to have cancer may receive inventive treatment at any time.
  • disclosed nanoparticles may be used to inhibit the growth of cancer cells, e.g., prostate cancer cells.
  • cancer cells e.g., prostate cancer cells.
  • the term “inhibits growth of cancer cells” or “inhibiting growth of cancer cells” refers to any slowing of the rate of cancer cell proliferation and/or migration, arrest of cancer cell proliferation and/or migration, or killing of cancer cells, such that the rate of cancer cell growth is reduced in comparison with the observed or predicted rate of growth of an untreated control cancer cell.
  • the term “inhibits growth” can also refer to a reduction in size or disappearance of a cancer cell or tumor, as well as to a reduction in its metastatic potential.
  • such an inhibition at the cellular level may reduce the size, deter the growth, reduce the aggressiveness, or prevent or inhibit metastasis of a cancer in a patient.
  • suitable indicia may be any of a variety of suitable indicia, whether cancer cell growth is inhibited.
  • Inhibition of cancer cell growth may be evidenced, for example, by arrest of cancer cells in a particular phase of the cell cycle, e.g., arrest at the G2/M phase of the cell cycle. Inhibition of cancer cell growth can also be evidenced by direct or indirect measurement of cancer cell or tumor size. In human cancer patients, such measurements generally are made using well known imaging methods such as magnetic resonance imaging, computerized axial tomography and X-rays. Cancer cell growth can also be determined indirectly, such as by determining the levels of circulating carcinoembryonic antigen, prostate specific antigen or other cancer-specific antigens that are correlated with cancer cell growth. Inhibition of cancer growth is also generally correlated with prolonged survival and/or increased health and well-being of the subject.
  • the synthesis is accomplished by ring opening polymerization of d,l-lactide with ⁇ -hydroxy- ⁇ -methoxypoly(ethylene glycol) as the macro-initiator, and performed at an elevated temperature using Tin (II) 2-Ethyl hexanoate as a catalyst, as shown below (PEG Mn ⁇ 5,000 Da; PLA Mn ⁇ 16,000 Da; PEGPLA M n ⁇ 21,000 Da)
  • the polymer is purified by dissolving the polymer in dichloromethane, and precipitating it in a mixture of hexane and diethyl ether.
  • the polymer recovered from this step shall be dried in an oven.
  • An organic phase is formed composed of a mixture of sirolimus and polymer (homopolymer, co-polymer, and co-polymer with ligand).
  • the organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent.
  • aqueous phase is composed of a surfactant and some dissolved solvent.
  • about 30% solids in the organic phase is used.
  • the primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer.
  • the rotor/stator yielded a homogeneous milky solution, while the stir bar produced a visibly larger coarse emulsion. It was observed that the stir bar method resulted in significant oil phase droplets adhering to the side of the feed vessel, suggesting that while the coarse emulsion size is not a process parameter critical to quality, it should be made suitably fine in order to prevent yield loss or phase separation. Therefore the rotor stator is used as the standard method of coarse emulsion formation, although a high speed mixer may be suitable at a larger scale.
  • the primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer.
  • the size of the coarse emulsion does not significantly affect the particle size after successive passes (1-3) through the homogenizer.
  • M-110-EH ( FIG. 3 ).
  • the standard operating pressure used for the M-110EH is 4000-5000 psi per interaction chamber, which is the minimum processing pressure on the unit.
  • the M-110EH also has the option of one or two interaction chambers. It comes standard with a restrictive Y-chamber, in series with a less restrictive 200 ⁇ m Z-chamber. It was found that the particle size was actually reduced when the Y-chamber was removed and replaced with a blank chamber. Furthermore, removing the Y-chamber significantly increases the flow rate of emulsion during processing.
  • Table A summarizes the emulsification process parameters.
  • the fine emulsion is then quenched by addition to deionized water at a given temperature under mixing.
  • the emulsion is added to a cold aqueous quench under agitation. This serves to extract a significant portion of the oil phase solvents, effectively hardening the nanoparticles for downstream filtration. Chilling the quench significantly improved drug encapsulation.
  • the quench:emulsion ratio is approximately 5:1.
  • Tween 80 A solution of 35% (wt %) of Tween 80 is added to the quench to achieve approximately 2% Tween 80 overall After the emulsion is quenched a solution of Tween-80 is added which acts as a drug solubilizer, allowing for effective removal of unencapsulated drug during filtration. Table B indicates each of the quench process parameters.
  • the temperature must remain cold enough with a dilute enough suspension (low enough concentration of solvents) to remain below the T g of the particles. If the Q:E ratio is not high enough, then the higher concentration of solvent plasticizes the particles and allows for drug leakage. Conversely, colder temperatures allow for high drug encapsulation at low Q:E ratios (to ⁇ 3:1), making it possible to run the process more efficiently.
  • the nanoparticles are then isolated through a tangential flow filtration process to concentrate the nanoparticle suspension and buffer exchange the solvents, free drug, and drug solubilizer from the quench solution into water.
  • a regenerated cellulose membrane is used with a molecular weight cutoffs (MWCO) of 300.
  • a constant volume diafiltration (DF) is performed to remove the quench solvents, free drug and Tween-80.
  • DF constant volume diafiltration
  • buffer is added to the retentate vessel at the same rate the filtrate is removed.
  • Crossflow rate refers to the rate of the solution flow through the feed channels and across the membrane. This flow provides the force to sweep away molecules that can foul the membrane and restrict filtrate flow.
  • the transmembrane pressure is the force that drives the permeable molecules through the membrane.
  • Diafiltration Concentration of 30 mg/ml Diafiltration is most efficient at [NP] ⁇ 50 mg/ml with Nanoparticle open channel TFF membranes based on flux rates and Suspension for throughput. With coarse channel membranes the flux rate Diafiltration is optimized at ⁇ 30 mg/ml in the starting buffer. Number of ⁇ 15 (based on About 15 diavolumes are needed to effectively remove Diavolumes flux increase) tween-80. End point of diafiltration is determined by in- process control (flux increase plateau). Membrane Area ⁇ 1 m 2 /kg Membranes sized based on anticipated flux rates and volumes required.
  • the filtered nanoparticle slurry is then thermal cycled to an elevated temperature during workup.
  • a small portion typically 5-10% of the encapsulated drug is released from the nanoparticles very quickly after its first exposure to 25° C. Because of this phenomenon, batches that are held cold during the entire workup are susceptible to free drug or drug crystals forming during delivery or any portion of unfrozen storage.
  • this ‘loosely encapsulated’ drug can be removed and improve the product stability at the expense of a small drop in drug loading. 5 diavolumes is used as the amount for cold processing prior to the 25° C. treatment.
  • the nanoparticle suspension is passed through a sterilizing grade filter (0.2 ⁇ m absolute).
  • Pre-filters are used to protect the sterilizing grade filter in order to use a reasonable filtration area/time for the process. Values are as summarized in Table D.
  • the filtration train is Ertel Alsop Micromedia XL depth filter M953P membrane (0.2 ⁇ m Nominal); Pall SUPRAcap with Seitz EKSP depth filter media (0.1-0.3 ⁇ m Nominal); Pall Life Sciences Supor EKV 0.65/0.2 micron sterilizing grade PES filter.
  • 0.2 m2 of filtration surface area per kg of nanoparticles for depth filters and 1.3 m2 of filtration surface area per kg of nanoparticles for the sterilizing grade filters can be used.
  • Sugar-based excipients sucrose, trehalose, or mannitol
  • sucrose trehalose, or mannitol
  • One formulation includes 10 wt % sucrose, which contains excess sucrose to what is required and is the same osmolality as physiological saline.
  • Table E shows that 16/5 PLA-PEG co-polymer is less susceptible to freeze-thaw aggregation.
  • An in vitro release method is used to determine the initial burst phase release from nanoparticles at both ambient and 37° C. conditions.
  • a dialysis system was designed. After obtaining an ultracentrifuge capable of pelleting 100 nm particles, the dialysis membranes were eliminated and centrifugation was used to separate released drug from encapsulated drug.
  • the dialysis system is as follows: 3 mL slurry of sirolimus nanoparticles (approx 250 ⁇ g/mL sirolimus PLGA/PLA nanoparticles, corresponding to 2.5 mg/mL solid concentration) in DI-water is placed into the inner tube of a 300 kDa MWCO dialyzer by pipetting. The nanoparticle is suspension in this media. The dialyzer is placed into a glass bottles containing 130 ml release media (2.5% hydroxyl beta cyclodextrin in PBS), which is continually stirred at 150 rpm using a shaker to prevent the formation of an unstirred water layer at the membrane/outer solution interface. At pre-determined time points, aliquot of samples (1 mL) were withdrawn from the outer solution (dialysate) and analyzed for sirolimus concentration by HPLC.
  • the centrifugal system is run using similar conditions at lower suspension volumes without dialysis bags. Samples are centrifuged at 60,000 g for 30 minutes and the supernatant is assayed for sirolimus content to measured released sirolimus.
  • DLS dynamic light scattering
  • laser diffraction is performed using a Brookhaven ZetaPals instrument at 25° C. in dilute aqueous suspension using a 660 nm laser scattered at 90° and analyzed using the Cumulants and NNLS methods (TP008).
  • Laser diffraction is performed with a Horiba LS950 instrument in dilute aqueous suspension using both a HeNe laser at 633 nm and an LED at 405 nm, scattered at 90° and analyzed using the Mie optical model (TP009).
  • the output from the DLS is associated with the hydrodynamic radius of the particles, which includes the PEG ‘corona’, while the laser diffraction instrument is more closely associated with the geometric size of the PLA particle ‘core’.
  • Nanoparticle batches were prepared using the general procedure of Example 2, with 80% (w/w) Polymer-PEG or Polymer-PEG with homopolymer PLA at 40% (w/w) each, with a batch of % total solids of 5%, 15% and 30%. Solvents used were: 21% benzyl alcohol and 79% ethyl acetate (w/w). For each 2 gram batch size, 400 mg of drug was used and 1.6 g of 16-5 Polymer-PEG or 0.8 g of 16-5 Polymer-PEG+0.8 g of 10 kDa PLA (homopolymer) was used.
  • the organic phase (drug and polymer) is prepared in 2 g batches: To 20 mL scintillation vial add drug and polymer(s). The mass of solvents needed at % solids concentration is shown below:
  • An aqueous solution is prepared with 0.5% sodium cholate, 2% benzyl alcohol, and 4% ethyl acetate in water.
  • To a 2 L bottle add 7.5 g sodium cholate, 1402.5 g of DI water, 30 g of benzyl alcohol and 60 g of ethyl acetate, and mix on stir plate until dissolved.
  • a ratio of aqueous phase to oil phase of 5:1 is used.
  • the organic phase is poured into the aqueous solution and homogenized using IKA for 10 seconds at room temperature to form course emulsion.
  • the solution is fed through the homogenizer (110S) at 9 Kpsi (45 psi on gauge) for 2 discreet passes to form nanoemulsion.
  • the emulsion is poured into quench (D.I. water) at ⁇ 5 C while stirring on stir plate. Ratio of quench to emulsion is 8:1. 35% (w/w) Tween 80 is added in water to quench at ratio of 25:1 Tween 80 to drug.
  • the nanoparticles are concentrated through TFF and the quench is concentrated on TFF with 500 kDa Pall cassette (2 membrane) to ⁇ 100 mL. Diafiltering is used using ⁇ 20 diavolumes (2 liter) of cold DI water, and the volume is brought down to minimal volume then collect final slurry, ⁇ 100 mL.
  • the solids concentration of unfiltered final slurry is determined by the using tared 20 mL scintillation vial and adding 4 mL final slurry and dry under vacuum on lyo/oven and the weight of nanoparticles in the 4 mL of slurry dried down is determined.
  • Concentrated sucrose (0.666 g/g) is added to final slurry sample to attain 10% sucrose.
  • Solids concentration of 0.45 um filtered final slurry was determined by filtering about 5 mL of final slurry sample before addition of sucrose through 0.45 ⁇ m syringe filter; to tared 20 mL scintillation vial add 4 mL of filtered sample and dry under vacuum on lyo/oven.
  • Rapamycin (sirolimus) formulations The remaining sample of unfiltered final slurry was frozen with sucrose. Rapamycin (sirolimus) formulations:
  • Nanoparticles were prepared as in Example 2 and 6, except temsirolimus was used with 30% solid content in the organic phase before emulsion:
  • FIG. 9 depicts the weight % of temsirolimus and FIG. 10 depicts the nanoparticle for the different polymeric nanoparticles having temsirolimus.
  • the results of an in-vitro release experiment as in Example 6 shows the slow and sustained release of temsirolimus showed slow and sustained release, as shown in FIG. 11 .

Abstract

The present disclosure generally relates to therapeutic nanoparticles. Exemplary nanoparticles disclosed herein may include about 1 to about 20 weight percent of a mTOR inhibitor; and about 70 to about 99 weight percent biocompatible polymer.

Description

RELATED APPLICATIONS
This application claims priority to U.S. Ser. No. 61/061,760, filed Jun. 16, 2008; U.S. Ser. No. 61/105,916, filed Oct. 16, 2008, U.S. Ser. No. 61/106,777, filed Oct. 20, 2008; U.S. Ser. No. 61/169,514, filed Apr. 15, 2009; U.S. Ser. No. 61/175,209, filed May 4, 2009; U.S. Ser. No. 61/061,704, filed Jun. 16, 2008; U.S. Ser. No. 61/169,519, filed Apr. 15, 2009; U.S. Ser. No. 61/175,219 filed May 4, 2009; U.S. Ser. No. 61/061,697, filed Jun. 16, 2008; U.S. Ser. No. 61/088,159, filed Aug. 12, 2008; U.S. Ser. No. 61/169,541, filed Apr. 15, 2009; U.S. Ser. No. 61/175,226, filed May 4, 2009; U.S. Ser. No. 61/173,790, filed Apr. 29, 2009; each of which is hereby incorporated by reference in their entirety.
This invention was made with United States Government support under Cooperative Agreement Number 70NANB7H7021 awarded by the National Institute of Standard and Technology (NIST). The United States Government has certain rights in the Invention.
BACKGROUND
Systems that deliver certain drugs to a patient (e.g., targeted to a particular tissue or cell type or targeted to a specific diseased tissue but not normal tissue), or that control release of drugs has long been recognized as beneficial. For example, therapeutics that include an active drug and that are capable of locating in a particular tissue or cell type e.g., a specific diseased tissue, may reduce the amount of the drug in tissues of the body that do not require treatment. This is particularly important when treating a condition such as cancer where it is desirable that a cytotoxic dose of the drug is delivered to cancer cells without killing the surrounding non-cancerous tissue. Further, such therapeutics may reduce the undesirable and sometimes life threatening side effects common in anticancer therapy. For example, nanoparticle therapeutics may, due the small size, evade recognition within the body allowing for targeted and controlled delivery while e.g., remaining stable for an effective amount of time.
Therapeutics that offer such therapy and/or controlled release and/or targeted therapy also must be able to deliver an effective amount of drug. It can be a challenge to prepare nanoparticle systems that have an appropriate amount of drug associated each nanoparticle, while keeping the size of the nanoparticles small enough to have advantageous delivery properties. For example, while it is desirable to load a nanoparticle with a high quantity of therapeutic agent, nanoparticle preparations that use a drug load that is too high will result in nanoparticles that are too large for practical therapeutic use. Further, it may be desirable for therapeutic nanoparticles to remain stable so as to e.g. substantially limit rapid or immediate release of the therapeutic agent.
Accordingly, a need exists for new nanoparticle formulations and methods of making such nanoparticles and compositions, that can deliver therapeutic levels of drugs to treat diseases such as cancer, while also reducing patient side effects.
SUMMARY
In one aspect, the invention provides therapeutic nanoparticle that includes an active agent or therapeutic agent, e.g. an mTOR inhibitor or pharmaceutically acceptable salts thereof, and one, two, or three biocompatible polymers. For example, disclosed herein is a therapeutic nanoparticle comprising about 1 to about 20 weight percent of a therapeutic agent (such as for example sirolimus, temsirolimus, or everolimus) and about 50 to about 99 weight percent of a biocompatible polymer, e.g. about 70 to about 99 weight percent of a biocompatible polymer. For example, the biocompatible polymer may be a diblock poly(lactic) acid-poly(ethylene)glycol copolymer (e.g. PLA-PEG) or a diblock (poly(lactic)-co-poly (glycolic) acid)-poly(ethylene)glycol copolymer (e.g. PLGA-PEG), or the biocompatible polymer may two or more biocompatible polymers, for example, the therapeutic nanoparticles can also include a homopolymer such as poly(lactic) acid homopolymer. For example, a disclosed therapeutic nanoparticle may include about 1 to about 20 weight percent, e.g about 2 to about 20 weight percent or about 10 to about 20 weight percent, of a mTOR inhibitor; and about 70 to about 99 weight percent biocompatible polymer, wherein the biocompatible polymer is selected from the group consisting of a) a diblock poly(lactic) acid-poly(ethylene)glycol copolymer, b) a diblock poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer, c) a combination of a) or b) and a poly (lactic) acid homopolymer or poly(lactic)-co-(glycolic) acid; and d) a combination of a) or b) and a poly (lactic) acid homopolymer or poly(lactic)-co-(glycolic) acid.
The diameter of disclosed nanoparticles may be, for example, about 60 to about 120 nm, or about 70 to about 120 nm. Disclosed therapeutic nanoparticles may be stable for at least 5 days at 25° C., e.g. may remain stable over 5 days in vitro, e.g. in a sucrose solution. In another embodiment, disclosed particles may substantially immediately release less than about 2% or less than about 5%, or even less than about 10% of the therapeutic agent when placed in a phosphate buffer solution at room temperature, or at 37° C. For example, disclosed nanoparticles may substantially retain the therapeutic agent for at least 5 days at 25° C. In another embodiment, disclosed nanoparticles may release the therapeutic agent over a period of at least 1 day or more when administered to a patient.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is flow chart for an emulsion process for forming disclosed nanoparticle.
FIG. 2 is a flow diagram for a disclosed emulsion process.
FIG. 3 depicts the effect of coarse emulsion preparation on quenched particle size. Placebo organic at 30% solids was used, emulsified at 5:1 W:O using standard aqueous phase (1% sodium cholate, 2% benzyl alcohol, 4% ethyl acetate).
FIG. 4 depicts the effect of feed pressure on resultant particle size.
FIG. 5 depicts the particle size dependence on scale. Placebo organic phase consisted of 25.5% polymer stock of 50:50 16.5/5 PLA/PEG:8.2 PLA. Organic phase was emulsified 5:1 O:W with standard aqueous phase, and multiple discreet passes were performed, quenching a small portion of emulsion after each pass. The indicated scale represents the total solids of the formulation.
FIG. 6 depicts the effect of solids concentration on particle size.
FIG. 7 depicts the effect of solids concentration and poly(lactic) homopolymer on loading percentage of sirolimus (rapamycin).
FIG. 8 depicts in vitro release of sirolimus over time for disclosed nanoparticles
FIG. 9 depicts the effects of poly(lactic) homopolymer on loading percentage of temsirolimus.
FIG. 10 depicts the effect of solids concentration on particle size of temsirolimus containing particles.
FIG. 11 depicts in vitro release of temsirolimus over time for disclosed nanoparticles
DETAILED DESCRIPTION
The present invention generally relates to polymeric nanoparticles that include an active or therapeutic agent or drug, and methods of making and using such therapeutic nanoparticles. In general, a “nanoparticle” refers to any particle having a diameter of less than 1000 nm, e.g. about 10 nm to about 200 nm. Disclosed therapeutic nanoparticles may include nanoparticles having a diameter of about 60 to about 120 nm, or about 70 to about 130 nm, or about 60 to about 140 nm, or about 70 nm to about 140 nm.
Disclosed nanoparticles may include about 0.2 to about 35 weight percent, about 3 to about 40 weight percent, about 5 to about 30 weight percent, about 1 to about 20 weight percent, about 10 to about 30 weight percent, about 5 to about 15 percent, about 15 to 25 weight percent, or even about 4 to about 25 weight percent, e.g. about 10 weight percent of an active agent, such as antineoplastic agent, e.g. a mTOR inhibiting agent (for example sirolimus, temsirolimus or everolimus).
Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers. For example, a contemplated nanoparticle may include about 60 to about 99 weight percent of one, two, three or more biocompatible polymers such as one or more co-polymers (e.g. a diblock polymer) that include a biodegradable polymer (for example poly(lactic)acid and polyethylene glycol, and optionally about 0 to about 50 weight percent of a homopolymer, e.g. biodegradable polymer such as poly(lactic) acid.
Polymers
In some embodiments, disclosed nanoparticles include a matrix of polymers. Disclosed nanoparticles may include one or more polymers, e.g. a diblock co-polymer and/or a monopolymer. Disclosed therapeutic nanoparticles include a therapeutic agent can may associated with the surface of, encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix.
A wide variety of polymers and methods for forming particles therefrom are known in the art of drug delivery. In some embodiments, the disclosure is directed toward nanoparticles with at least one polymer, for example, a first polymer that may be a co-polymer, e.g. a diblock co-polymer, and optionally a polymer that may be for example a homopolymer.
Any polymer can be used in accordance with the present invention. Polymers can be natural or unnatural (synthetic) polymers. Polymers can be homopolymers or copolymers comprising two or more monomers. In terms of sequence, copolymers can be random, block, or comprise a combination of random and block sequences. Contemplated polymers may be biocompatible and/or biodegradable.
The term “polymer,” as used herein, is given its ordinary meaning as used in the art, i.e., a molecular structure comprising one or more repeat units (monomers), connected by covalent bonds. The repeat units may all be identical, or in some cases, there may be more than one type of repeat unit present within the polymer. In some cases, the polymer can be biologically derived, i.e., a biopolymer. Non-limiting examples include peptides or proteins. In some cases, additional moieties may also be present in the polymer, for example biological moieties such as those described below. If more than one type of repeat unit is present within the polymer, then the polymer is said to be a “copolymer.” It is to be understood that in any embodiment employing a polymer, the polymer being employed may be a copolymer in some cases. The repeat units forming the copolymer may be arranged in any fashion. For example, the repeat units may be arranged in a random order, in an alternating order, or as a block copolymer, i.e., comprising one or more regions each comprising a first repeat unit (e.g., a first block), and one or more regions each comprising a second repeat unit (e.g., a second block), etc. Block copolymers may have two (a diblock copolymer), three (a triblock copolymer), or more numbers of distinct blocks.
Disclosed particles can include copolymers, which, in some embodiments, describes two or more polymers (such as those described herein) that have been associated with each other, usually by covalent bonding of the two or more polymers together. Thus, a copolymer may comprise a first polymer and a second polymer, which have been conjugated together to form a block copolymer where the first polymer can be a first block of the block copolymer and the second polymer can be a second block of the block copolymer. Of course, those of ordinary skill in the art will understand that a block copolymer may, in some cases, contain multiple blocks of polymer, and that a “block copolymer,” as used herein, is not limited to only block copolymers having only a single first block and a single second block. For instance, a block copolymer may comprise a first block comprising a first polymer, a second block comprising a second polymer, and a third block comprising a third polymer or the first polymer, etc. In some cases, block copolymers can contain any number of first blocks of a first polymer and second blocks of a second polymer (and in certain cases, third blocks, fourth blocks, etc.). In addition, it should be noted that block copolymers can also be formed, in some instances, from other block copolymers. For example, a first block copolymer may be conjugated to another polymer (which may be a homopolymer, a biopolymer, another block copolymer, etc.), to form a new block copolymer containing multiple types of blocks, and/or to other moieties (e.g., to non-polymeric moieties).
In some embodiments, the polymer (e.g., copolymer, e.g., block copolymer) can be amphiphilic, i.e., having a hydrophilic portion and a hydrophobic portion, or a relatively hydrophilic portion and a relatively hydrophobic portion. A hydrophilic polymer can be one generally that attracts water and a hydrophobic polymer can be one that generally repels water. A hydrophilic or a hydrophobic polymer can be identified, for example, by preparing a sample of the polymer and measuring its contact angle with water (typically, the polymer will have a contact angle of less than 60°, while a hydrophobic polymer will have a contact angle of greater than about 60°). In some cases, the hydrophilicity of two or more polymers may be measured relative to each other, i.e., a first polymer may be more hydrophilic than a second polymer. For instance, the first polymer may have a smaller contact angle than the second polymer.
In one set of embodiments, a polymer (e.g., copolymer, e.g., block copolymer) contemplated herein includes a biocompatible polymer, i.e., the polymer that does not typically induce an adverse response when inserted or injected into a living subject, for example, without significant inflammation and/or acute rejection of the polymer by the immune system, for instance, via a T-cell response. Accordingly, the therapeutic particles contemplated herein can be non-immunogenic. The term non-immunogenic as used herein refers to endogenous growth factor in its native state which normally elicits no, or only minimal levels of, circulating antibodies, T-cells, or reactive immune cells, and which normally does not elicit in the individual an immune response against itself.
Biocompatibility typically refers to the acute rejection of material by at least a portion of the immune system, i.e., a nonbiocompatible material implanted into a subject provokes an immune response in the subject that can be severe enough such that the rejection of the material by the immune system cannot be adequately controlled, and often is of a degree such that the material must be removed from the subject. One simple test to determine biocompatibility can be to expose a polymer to cells in vitro; biocompatible polymers are polymers that typically will not result in significant cell death at moderate concentrations, e.g., at concentrations of 50 micrograms/106 cells. For instance, a biocompatible polymer may cause less than about 20% cell death when exposed to cells such as fibroblasts or epithelial cells, even if phagocytosed or otherwise uptaken by such cells. Non-limiting examples of biocompatible polymers that may be useful in various embodiments of the present invention include polydioxanone (PDO), polyhydroxyalkanoate, polyhydroxybutyrate, poly(glycerol sebacate), polyglycolide, polylactide, PLGA, polycaprolactone, or copolymers or derivatives including these and/or other polymers.
In certain embodiments, contemplated biocompatible polymers may be biodegradable, i.e., the polymer is able to degrade, chemically and/or biologically, within a physiological environment, such as within the body. As used herein, “biodegradable” polymers are those that, when introduced into cells, are broken down by the cellular machinery (biologically degradable) and/or by a chemical process, such as hydrolysis, (chemically degradable) into components that the cells can either reuse or dispose of without significant toxic effect on the cells. In one embodiment, the biodegradable polymer and their degradation byproducts can be biocompatible.
For instance, a contemplated polymer may be one that hydrolyzes spontaneously upon exposure to water (e.g., within a subject), the polymer may degrade upon exposure to heat (e.g., at temperatures of about 37° C.). Degradation of a polymer may occur at varying rates, depending on the polymer or copolymer used. For example, the half-life of the polymer (the time at which 50% of the polymer can be degraded into monomers and/or other non-polymeric moieties) may be on the order of days, weeks, months, or years, depending on the polymer. The polymers may be biologically degraded, e.g., by enzymatic activity or cellular machinery, in some cases, for example, through exposure to a lysozyme (e.g., having relatively low pH). In some cases, the polymers may be broken down into monomers and/or other non-polymeric moieties that cells can either reuse or dispose of without significant toxic effect on the cells (for example, polylactide may be hydrolyzed to form lactic acid, polyglycolide may be hydrolyzed to form glycolic acid, etc.).
In some embodiments, polymers may be polyesters, including copolymers comprising lactic acid and glycolic acid units, such as poly(lactic acid-co-glycolic acid) and poly(lactide-co-glycolide), collectively referred to herein as “PLGA”; and homopolymers comprising glycolic acid units, referred to herein as “PGA,” and lactic acid units, such as poly-L-lactic acid, poly-D-lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as “PLA.” In some embodiments, exemplary polyesters include, for example, polyhydroxyacids; PEGylated polymers and copolymers of lactide and glycolide (e.g., PEGylated PLA, PEGylated PGA, PEGylated PLGA, and derivatives thereof. In some embodiments, polyesters include, for example, polyanhydrides, poly(ortho ester) PEGylated poly(ortho ester), poly(caprolactone), PEGylated poly(caprolactone), polylysine, PEGylated polylysine, poly(ethylene imine), PEGylated poly(ethylene imine), poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[α-(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
In some embodiments, a polymer may be PLGA. PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA can be characterized by the ratio of lactic acid:glycolic acid. Lactic acid can be L-lactic acid, D-lactic acid, or D,L-lactic acid. The degradation rate of PLGA can be adjusted by altering the lactic acid-glycolic acid ratio. In some embodiments, PLGA to be used in accordance with the present invention can be characterized by a lactic acid:glycolic acid molar ratio of approximately 85:15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
In some embodiments, the ratio of lactic acid to glycolic acid monomers in the polymer of the particle (e.g., the PLGA block copolymer or PLGA-PEG block copolymer), may be selected to optimize for various parameters such as water uptake, therapeutic agent release and/or polymer degradation kinetics can be optimized.
In some embodiments, polymers may be one or more acrylic polymers. In certain embodiments, acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid polyacrylamide, amino alkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers. The acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.
In some embodiments, polymers can be cationic polymers. In general, cationic polymers are able to condense and/or protect negatively charged strands of nucleic acids (e.g. DNA, RNA, or derivatives thereof). Amine-containing polymers such as poly(lysine), polyethylene imine (PEI), and poly(amidoamine) dendrimers are contemplated for use, in some embodiments, in a disclosed particle.
In some embodiments, polymers can be degradable polyesters bearing cationic side chains. Examples of these polyesters include poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester). A polymer (e.g., copolymer, e.g., block copolymer) containing poly(ethylene glycol) repeat units can also be referred to as a “PEGylated” polymer. Such polymers can control inflammation and/or immunogenicity (i.e., the ability to provoke an immune response) and/or lower the rate of clearance from the circulatory system via the reticuloendothelial system (RES), due to the presence of the poly(ethylene glycol) groups.
PEGylation may also be used, in some cases, to decrease charge interaction between a polymer and a biological moiety, e.g., by creating a hydrophilic layer on the surface of the polymer, which may shield the polymer from interacting with the biological moiety. In some cases, the addition of poly(ethylene glycol) repeat units may increase plasma half-life of the polymer (e.g., copolymer, e.g., block copolymer), for instance, by decreasing the uptake of the polymer by the phagocytic system while decreasing transfection/uptake efficiency by cells. Those of ordinary skill in the art will know of methods and techniques for PEGylating a polymer, for example, by using EDC (1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride) and NHS (N-hydroxysuccinimide) to react a polymer to a PEG group terminating in an amine, by ring opening polymerization techniques (ROMP), or the like.
Particles disclosed herein may or may not contain PEG. In addition, certain embodiments can be directed towards copolymers containing poly(ester-ether)s, e.g., polymers having repeat units joined by ester bonds (e.g., R—C(O)—O—R′ bonds) and ether bonds (e.g., R—O—R′ bonds). In some embodiments of the invention, a biodegradable polymer, such as a hydrolyzable polymer, containing carboxylic acid groups, may be conjugated with poly(ethylene glycol) repeat units to form a poly(ester-ether).
It is contemplated that PEG may include a terminal end group, for example, when PEG is not conjugated to a ligand. For example, PEG may terminate in a hydroxyl, a methoxy or other alkoxyl group, a methyl or other alkyl group, an aryl group, a carboxylic acid, an amine, an amide, an acetyl group, a guanidino group, or an imidazole. Other contemplated end groups include azide, alkyne, maleimide, aldehyde, hydrazide, hydroxylamine, alkoxyamine, or thiol moieties.
In one embodiment, the molecular weight of the polymers can be optimized for effective treatment as disclosed herein. For example, the molecular weight of a polymer may influence particle degradation rate (such as when the molecular weight of a biodegradable polymer can be adjusted), solubility, water uptake, and drug release kinetics. For example, the molecular weight of the polymer can be adjusted such that the particle biodegrades in the subject being treated within a reasonable period of time (ranging from a few hours to 1-2 weeks, 3-4 weeks, 5-6 weeks, 7-8 weeks, etc.). A disclosed particle can for example comprise a copolymer of PEG and PLGA, the PEG can have a molecular weight of 1,000-20,000, e.g., 5,000-20,000, e.g., 10,000-20,000, and the PLGA can have a molecular weight of 5,000-100,000, e.g., 20,000-70,000, e.g., 20,000-50,000.
For example, disclosed here is an exemplary therapeutic nanoparticle that includes about 10 to about 99 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer, or about 20 to about 80 weight percent, about 40 to about 80 weight percent, or about 30 to about 50 weight percent, or about 70 to about 90 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or poly(lactic)-co-poly (glycolic) acid-poly(ethylene)glycol copolymer. Exemplary poly(lactic) acid-poly(ethylene)glycol copolymers can include a number average molecular weight of about 15 to about 20 kDa, or about 10 to about 25 kDa of poly(lactic) acid and a number average molecular weight of about 4 to about 6, or about 2 kDa to about 10 kDa of poly(ethylene)glycol.
Disclosed nanoparticles may optionally include about 1 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid (which does not include PEG, e.g a homopolymer of PLA), or may optionally include about 1 to about 50 weight percent, or about 10 to about 50 weight percent or about 30 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid. For example, poly(lactic) or poly(lactic)-co-poly(glycolic) acid may have a number average molecule weight of about 5 to about 15 kDa, or about 5 to about 12 kDa. Exemplary homopolymeric PLA may have a number average molecular weight of about 5 to about 10 kDa. Exemplary PLGA may have a number average molecular weight of about 8 to about 12 kDa.
In certain embodiments, disclosed polymers of may be conjugated to a lipid, e.g. “end-capped,” for example, may include a lipid-terminated PEG. As described below, the lipid portion of the polymer can be used for self assembly with another polymer, facilitating the formation of a nanoparticle. For example, a hydrophilic polymer could be conjugated to a lipid that will self assemble with a hydrophobic polymer.
Exemplary lipids include fatty acids such as long chain (e.g., C8-C50), substituted or unsubstituted hydrocarbons. In some embodiments, a fatty acid group can be a C10-C20 fatty acid or salt thereof. In some embodiments, a fatty acid group can be a C15-C20 fatty acid or salt thereof. In some embodiments, a fatty acid can be unsaturated, monounsaturated, or polyunsaturated. For example, a fatty acid group can be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid. In some embodiments, a fatty acid group can be one or more of palmitoleic, oleic, vaccenic, linoleic, alpha-linolenic, gamma-linoleic, arachidonic, gadoleic, arachidonic, eicosapentaenoic, docosahexaenoic, or erucic acid.
In a particular embodiment, the lipid is of the Formula V:
Figure US08613951-20131224-C00001

and salts thereof, wherein each R is, independently, C1-30 alkyl. In one embodiment of Formula V, the lipid is 1,2 distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), and salts thereof, e.g., the sodium salt.
In one embodiment, optional small molecule targeting moieties are bonded, e.g., covalently bonded, to the lipid component of the nanoparticle. For example, contemplated herein is also a nanoparticle comprising a therapeutic agent, a polymeric matrix comprising functionalized and non-functionalized polymers, a lipid, and a low-molecular weight targeting ligand, wherein the targeting ligand is bonded, e.g., covalently bonded, to the lipid component of the nanoparticle.
Targeting Moieties
Provided herein are nanoparticles that may include an optional targeting moiety, i.e., a moiety able to bind to or otherwise associate with a biological entity, for example, a membrane component, a cell surface receptor, prostate specific membrane antigen, or the like. A targeting moiety present on the surface of the particle may allow the particle to become localized at a particular targeting site, for instance, a tumor, a disease site, a tissue, an organ, a type of cell, etc. The drug or other payload may then, in some cases, be released from the particle and allowed to interact locally with the particular targeting site.
In one embodiment of the instant invention, the targeting moiety may be a low-molecular weight ligand, e.g., a low-molecular weight PSMA ligand. For example, a targeting portion may cause the particles to become localized to a tumor, a disease site, a tissue, an organ, a type of cell, etc. within the body of a subject, depending on the targeting moiety used. For example, a low-molecular weight PSMA ligand may become localized to prostate cancer cells. The subject may be a human or non-human animal. Examples of subjects include, but are not limited to, a mammal such as a dog, a cat, a horse, a donkey, a rabbit, a cow, a pig, a sheep, a goat, a rat, a mouse, a guinea pig, a hamster, a primate, a human or the like.
Contemplated targeting moieties include small molecules. In certain embodiments, the term “small molecule” refers to organic compounds, whether naturally-occurring or artificially created (e.g., via chemical synthesis) that have relatively low molecular weight and that are not proteins, polypeptides, or nucleic acids. Small molecules typically have multiple carbon-carbon bonds. In certain embodiments, small molecules are less than about 2000 g/mol in size. In some embodiments, small molecules are less than about 1500 g/mol or less than about 1000 g/mol. In some embodiments, small molecules are less than about 800 g/mol or less than about 500 g/mol, for example about 100 g/mol to about 600 g/mol, or about 200 g/mol to about 500 g/mol. For example, a ligand may be a the low-molecular weight PSMA ligand such as
Figure US08613951-20131224-C00002

and enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof.
In some embodiments, small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include PSMA peptidase inhibitors such as 2-PMPA, GPI5232, VA-033, phenylalkylphosphonamidates and/or analogs and derivatives thereof. In some embodiments, small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include thiol and indole thiol derivatives, such as 2-MPPA and 3-(2-mercaptoethyl)-1H-indole-2-carboxylic acid derivatives. In some embodiments, small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include hydroxamate derivatives. In some embodiments, small molecule targeting moieties that may be used to target cells associated with prostate cancer tumors include PBDA- and urea-based inhibitors, such as ZJ 43, ZJ 11, ZJ 17, ZJ 38 and/or and analogs and derivatives thereof, androgen receptor targeting agents (ARTAs), polyamines, such as putrescine, spermine, and spermidine, inhibitors of the enzyme glutamate carboxylase II (GCPII), also known as NAAG Peptidase or NAALADase.
In another embodiment of the instant invention, the targeting moiety can be a ligand that targets Her2, EGFR, or toll receptors. For example, contemplated the targeting moieties may include a nucleic acid, polypeptide, glycoprotein, carbohydrate, or lipid. For example, a targeting moiety can be a nucleic acid targeting moiety (e.g. an aptamer, e.g., the A10 aptamer) that binds to a cell type specific marker. In general, an aptamer is an oligonucleotide (e.g., DNA, RNA, or an analog or derivative thereof) that binds to a particular target, such as a polypeptide. In some embodiments, a targeting moiety may be a naturally occurring or synthetic ligand for a cell surface receptor, e.g., a growth factor, hormone, LDL, transferrin, etc. A targeting moiety can be an antibody, which term is intended to include antibody fragments, characteristic portions of antibodies, single chain targeting moieties can be identified, e.g., using procedures such as phage display. Targeting moieties may be a targeting peptide or targeting peptidomimetic has a length of up to about 50 residues. For example, a targeting moieties may include the amino acid sequence AKERC, CREKA, ARYLQKLN or AXYLZZLN, wherein X and Z are variable amino acids, or conservative variants or peptidomimetics thereof. In particular embodiments, the targeting moiety is a peptide that includes the amino acid sequence AKERC, CREKA, ARYLQKLN or AXYLZZLN, wherein X and Z are variable amino acids, and has a length of less than 20, 50 or 100 residues. The CREKA (Cys Arg Glu Lys Ala) peptide or a peptidomimetic thereof peptide or the octapeptide AXYLZZLN are also contemplated as targeting moieties, as well as peptides, or conservative variants or peptidomimetics thereof, that binds or forms a complex with collagen IV, or the targets tissue basement membrane (e.g., the basement membrane of a blood vessel), can be used as a targeting moiety.
Exemplary targeting moieties include peptides that target ICAM (intercellular adhesion molecule, e.g. ICAM-1).
Targeting moieties disclosed herein are typically conjugated to a disclosed polymer or copolymer (e.g. PLA-PEG), and such a polymer conjugate may form part of a disclosed nanoparticle. For example, a disclosed therapeutic nanoparticle may optionally include about 0.2 to about 10 weight percent of a PLA-PEG or PLGA-PEG, wherein the PEG is functionalized with a targeting ligand. Contemplated therapeutic nanoparticles may include, for example, about 0.2 to about 10 mole percent PLA-PEG-ligand or poly (lactic) acid co poly (glycolic) acid-PEG-ligand. For example, PLA-PEG-ligand may include a PLA with a number average molecular weight of about 10 kDa to about 20 kDa and PEG with a number average molecular weight of about 4,000 to about 8,000.
Nanoparticles
Disclosed nanoparticles may have a substantially spherical (i.e., the particles generally appear to be spherical), or non-spherical configuration. For instance, the particles, upon swelling or shrinkage, may adopt a non-spherical configuration. In some cases, the particles may include polymeric blends. For instance, a polymer blend may include a first co-polymer that includes polyethylene glycol and a second polymer.
Disclosed nanoparticles may have a characteristic dimension of less than about 1 micrometer, where the characteristic dimension of a particle is the diameter of a perfect sphere having the same volume as the particle. For example, the particle can have a characteristic dimension of the particle can be less than about 300 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, less than about 50 nm, less than about 30 nm, less than about 10 nm, less than about 3 nm, or less than about 1 nm in some cases. In particular embodiments, disclosed nanoparticles may have a diameter of about 70 nm-200 nm, or about 70 nm to about 180 nm, about 80 nm to about 130 nm, about 80 nm to about 120 nm.
In one set of embodiments, the particles can have an interior and a surface, where the surface has a composition different from the interior, i.e., there may be at least one compound present in the interior but not present on the surface (or vice versa), and/or at least one compound is present in the interior and on the surface at differing concentrations. For example, in one embodiment, a compound, such as a targeting moiety (i.e., a low-molecular weight ligand) of a polymeric conjugate of the present invention, may be present in both the interior and the surface of the particle, but at a higher concentration on the surface than in the interior of the particle, although in some cases, the concentration in the interior of the particle may be essentially nonzero, i.e., there is a detectable amount of the compound present in the interior of the particle.
In some cases, the interior of the particle is more hydrophobic than the surface of the particle. For instance, the interior of the particle may be relatively hydrophobic with respect to the surface of the particle, and a drug or other payload may be hydrophobic, and readily associates with the relatively hydrophobic center of the particle. The drug or other payload can thus be contained within the interior of the particle, which can shelter it from the external environment surrounding the particle (or vice versa). For instance, a drug or other payload contained within a particle administered to a subject will be protected from a subject's body, and the body may also be substantially isolated from the drug for at least a period of time.
For example, disclosed herein is a therapeutic polymeric nanoparticle comprising a first non-functionalized polymer; an optional second non-functionalized polymer; an optional functionalized polymer comprising a targeting moiety; and a therapeutic agent, In a particular embodiment, the first non-functionalized polymer is PLA, PLGA, or PEG, or copolymers thereof, e.g. a diblock co-polymer PLA-PEG. For example, exemplary nanoparticle may have a PEG corona with a density of about 0.065 g/cm3, or about 0.01 to about 0.10 g/cm3.
Disclosed nanoparticles may be stable (e.g. retain substantially all active agent) for example in a solution that may contain a saccharide, for at least about 3 days, about 4 days or at least about 5 days at room temperature, or at 25° C.
In some embodiments, disclosed nanoparticles may also include a fatty alcohol, which may increase the rate of drug release. For example, disclosed nanoparticles may include a C8-C30 alcohol such as cetyl alcohol, octanol, stearyl alcohol, arachidyl alcohol, docosonal, or octasonal.
Nanoparticles may have controlled release properties, e.g., may be capable of delivering an amount of active agent to a patient, e.g., to specific site in a patient, over an extended period of time, e.g. over 1 day, 1 week, or more. In some embodiments, disclosed nanoparticles substantially immediately releases (e.g. over about 1 minute to about 30 minutes) less than about 2%, less than about 4%, less than about 5%, or less than about 10% of an active agent (e.g. a taxane) agent, for example when places in a phosphate buffer solution at room temperature and/or at 37° C.
In one embodiment, the invention comprises a nanoparticle comprising 1) a polymeric matrix and 2) an amphiphilic compound or layer that surrounds or is dispersed within the polymeric matrix forming a continuous or discontinuous shell for the particle, An amphiphilic layer can reduce water penetration into the nanoparticle, thereby enhancing drug encapsulation efficiency and slowing drug release. Further, these amphipilic layer protected nanoparticles can provide therapeutic advantages by releasing the encapsulated drug and polymer at appropriate times.
As used herein, the term “amphiphilic” refers to a property where a molecule has both a polar portion and a non-polar portion. Often, an amphiphilic compound has a polar head attached to a long hydrophobic tail. In some embodiments, the polar portion is soluble in water, while the non-polar portion is insoluble in water. In addition, the polar portion may have either a formal positive charge, or a formal negative charge. Alternatively, the polar portion may have both a formal positive and a negative charge, and be a zwitterion or inner salt. Exemplary amphiphilic compound include, for example, one or a plurality of the following: naturally derived lipids, surfactants, or synthesized compounds with both hydrophilic and hydrophobic moieties.
Specific examples of amphiphilic compounds include, but are not limited to, phospholipids, such as 1,2 distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcholine (DBPC), ditricosanoylphosphatidylcholine (DTPC), and dilignoceroylphatidylcholine (DLPC), incorporated at a ratio of between 0.01-60 (weight lipid/w polymer), most preferably between 0.1-30 (weight lipid/w polymer). Phospholipids which may be used include, but are not limited to, phosphatidic acids, phosphatidyl cholines with both saturated and unsaturated lipids, phosphatidyl ethanolamines, phosphatidylglycerols, phosphatidylserines, phosphatidylinositols, lysophosphatidyl derivatives, cardiolipin, and β-acyl-y-alkyl phospholipids. Examples of phospholipids include, but are not limited to, phosphatidylcholines such as dioleoylphosphatidylcholine, dimyristoylphosphatidylcholine, dipentadecanoylphosphatidylcholine dilauroylphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcho-line (DBPC), ditricosanoylphosphatidylcholine (DTPC), dilignoceroylphatidylcholine (DLPC); and phosphatidylethanolamines such as dioleoylphosphatidylethanolamine or 1-hexadecyl-2-palmitoylglycerophos-phoethanolamine. Synthetic phospholipids with asymmetric acyl chains (e.g., with one acyl chain of 6 carbons and another acyl chain of 12 carbons) may also be used.
In a particular embodiment, an amphiphilic component may include lecithin, and/or in particular, phosphatidylcholine.
Preparation of Nanoparticles
Another aspect of the invention is directed to systems and methods of making disclosed nanoparticles. In some embodiments, using two or more different polymers (e.g., a copolymer such as a diblock copolymer and a homopolymer) properties of particles may be controlled.
In a particular embodiment, the methods described herein form nanoparticles that have a high amount of encapsulated therapeutic agent, for example, may include about 1 to about 40 weight percent, or about 1 to about 30 weight percent, e.g. about 10 to about 25 weight percent or about 5 to about 20 weight percent therapeutic agent.
In an embodiment, a nanoemulsion process is provided, such as the process represented in FIGS. 1 and 2. For example, a therapeutic agent, a first polymer (for example, PLA-PEG or PLGA-PEG) and a second polymer (e.g. (PL(G)A or PLA), with an organic solution to form a first organic phase. Such first phase may include about 5 to about 50% weight solids, e.g about 5 to about 40% solids, or about 10 to about 30% solids, e.g. about 10%, 15%, 20% solids. The first organic phase may be combined with a first aqueous solution to form a second phase. The organic solution can include, for example, acetonitrile, tetrahydrofuran, ethyl acetate, isopropyl alcohol, isopropyl acetate, dimethylformamide, methylene chloride, dichloromethane, chloroform, acetone, benzyl alcohol, Tween 80, Span 80, or the like, and combinations thereof. In an embodiment, the organic phase may include benzyl alcohol, ethyl acetate, and combinations thereof. The second phase can be between about 1 and 50 weight %, e.g., 5-40 weight %, solids. The aqueous solution can be water, optionally in combination with one or more of sodium cholate, ethyl acetate, and benzyl alcohol.
For example, the oil or organic phase may use solvent that is only partially miscible with the nonsolvent (water). Therefore, when mixed at a low enough ratio and/or when using water pre-saturated with the organic solvents, the oil phase remains liquid. The oil phase may bee emulsified into an aqueous solution and, as liquid droplets, sheared into nanoparticles using, for example, high energy dispersion systems, such as homogenizers or sonicators. The aqueous portion of the emulsion, otherwise known as the “water phase”, may be surfactant solution consisting of sodium cholate and pre-saturated with ethyl acetate and benzyl alcohol.
Emulsifying the second phase to form an emulsion phase may be performed in one or two emulsification steps. For example, a primary emulsion may be prepared, and then emulsified to form a fine emulsion. The primary emulsion can be formed, for example, using simple mixing, a high pressure homogenizer, probe sonicator, stir bar, or a rotor stator homogenizer. The primary emulsion may be formed into a fine emulsion through the use of e.g. probe sonicator or a high pressure homogenizer, e.g. by using 1, 2, 3 or more passes through a homogenizer. For example, when a high pressure homogenizer is used, the pressure used may be about 4000 to about 8000 psi, or about 4000 to about 5000 psi, e.g. 4000 or 5000 psi.
Either solvent evaporation or dilution may be needed to complete the extraction of the solvent and solidify the particles. For better control over the kinetics of extraction and a more scalable process, a solvent dilution via aqueous quench may be used. For example, the emulsion can be diluted into cold water to a concentration sufficient to dissolve all of the organic solvent to form a quenched phase. Quenching may be performed at least partially at a temperature of about 5° C. or less. For example, water used in the quenching may be at a temperature that is less that room temperature (e.g. about 0 to about 10° C., or about 0 to about 5° C.).
In some embodiments, not all of the therapeutic agent is encapsulated in the particles at this stage, and a drug solubilizer is added to the quenched phase to form a solubilized phase. The drug solubilizer may be for example, Tween 80, Tween 20, polyvinyl pyrrolidone, cyclodextran, sodium dodecyl sulfate, or sodium cholate. For example, Tween-80 may added to the quenched nanoparticle suspension to solubilize the free drug and prevent the formation of drug crystals. In some embodiments, a ratio of drug solubilizer to therapeutic agent is about 100:1 to about 10:1.
The solubilized phase may be filtered to recover the nanoparticles. For example, ultrafiltration membranes may be used to concentrate the nanoparticle suspension and substantially eliminate organic solvent, free drug, and other processing aids (surfactants). Exemplary filtration may be performed using a tangential flow filtration system. For example, by using a membrane with a pore size suitable to retain nanoparticles while allowing solutes, micelles, and organic solvent to pass, nanoparticles can be selectively separated. Exemplary membranes with molecular weight cut-offs of about 300-500 kDa (˜5-25 nm) may be used.
Diafiltration may be performed using a constant volume approach, meaning the diafiltrate (cold deionized water, e.g. about 0° C. to about 5° C., or 0 to about 10° C.) may added to the feed suspension at the same rate as the filtrate is removed from the suspension. In some embodiments, filtering may include a first filtering using a first temperature of about 0 to about 5° C., or 0° C. to about 10° C., and a second temperature of about 20° C. to about 30° C., or 15° C. to about 35° C. For example, filtering may include processing about 1 to about 6 diavolumes at about 0 to about 5° C., and processing at least one diavolume (e.g. about 1 to about 3 or about 1-2 diavolumes) at about 20° C. to about 30° C.
After purifying and concentrating the nanoparticle suspension, the particles may be passed through one, two or more sterilizing and/or depth filters, for example, using ˜0.2 μm depth pre-filter.
In exemplary embodiment of preparing nanoparticles, an organic phase is formed composed of a mixture of a therapeutic agent, e.g., sirolimus, and polymer (homopolymer, and co-polymer). The organic phase may be mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and optionally dissolved solvent. A primary emulsion may then formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer. The primary emulsion is then formed into a fine emulsion through the use of e.g. high pressure homogenizer. Such fine emulsion may then quenched by, e.g. addition to deionized water under mixing. An exemplary quench:emulsion ratio may be about approximately 8.5:1. A solution of Tween (e.g., Tween 80) can then be added to the quench to achieve e.g. approximately 2% Tween overall, which may serves to dissolve free, unencapsulated drug. Formed nanoparticles may then be isolated through either centrifugation or ultrafiltration/diafiltration.
Therapeutic Agents
According to the present invention, any agents including, for example, therapeutic agents (e.g. anti-cancer agents), diagnostic agents (e.g. contrast agents; radionuclides; and fluorescent, luminescent, and magnetic moieties), prophylactic agents (e.g. vaccines), and/or nutraceutical agents (e.g. vitamins, minerals, etc.) may be delivered by the disclosed nanoparticles. Exemplary agents to be delivered in accordance with the present invention include, but are not limited to, small molecules (e.g. cytotoxic agents), nucleic acids (e.g., siRNA, RNAi, and microRNA agents), proteins (e.g. antibodies), peptides, lipids, carbohydrates, hormones, metals, radioactive elements and compounds, drugs, vaccines, immunological agents, etc., and/or combinations thereof. In some embodiments, the agent to be delivered is an agent useful in the treatment of cancer (e.g., an anti-neoplastic agent).
In a particular embodiment, the drug may be released in a controlled release manner from the particle and allowed to interact locally with the particular patient site (e.g., a tumor). The term “controlled release” is generally meant to encompass release of a substance (e.g., a drug) at a selected site or otherwise controllable in rate, interval, and/or amount. Controlled release encompasses, but is not necessarily limited to, substantially continuous delivery, patterned delivery (e.g., intermittent delivery over a period of time that is interrupted by regular or irregular time intervals), and delivery of a bolus of a selected substance (e.g., as a predetermined, discrete amount if a substance over a relatively short period of time (e.g., a few seconds or minutes)).
The active agent or drug may be a therapeutic agent such as an mTOR (mammalian target of rapamycin) inhibitor such as sirolimus (rapamycin), temsirolimus, or everolimus, a taxane or diterpene derivative such as paclitaxel (or its derivatives such as DHA-paclitaxel or PG-paxlitaxel) or docetaxel. In another embodiment, the active agent or drug may be a vinca alkaloid such as vinorelbine, vinblastine, vincristine, or vindesine.
Pharmaceutical Formulations
Nanoparticles disclosed herein may be combined with pharmaceutical acceptable carriers to form a pharmaceutical composition. As would be appreciated by one of skill in this art, the carriers may be chosen based on the route of administration as described below, the location of the target issue, the drug being delivered, the time course of delivery of the drug, etc.
The pharmaceutical compositions and particles disclosed herein can be administered to a patient by any means known in the art including oral and parenteral routes. The term “patient,” as used herein, refers to humans as well as non-humans, including, for example, mammals, birds, reptiles, amphibians, and fish. For instance, the non-humans may be mammals (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a primate, or a pig). In certain embodiments parenteral routes are desirable since they avoid contact with the digestive enzymes that are found in the alimentary canal. According to such embodiments, inventive compositions may be administered by injection (e.g., intravenous, subcutaneous or intramuscular, intraperitoneal injection), rectally, vaginally, topically (as by powders, creams, ointments, or drops), or by inhalation (as by sprays).
In a particular embodiment, disclosed nanoparticles may be administered to a subject in need thereof systemically, e.g., by IV infusion or injection.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension, or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. In one embodiment, the inventive conjugate is suspended in a carrier fluid comprising 1% (w/v) sodium carboxymethyl cellulose and 0.1% (v/v) TWEEN™ 80. The injectable formulations can be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the encapsulated or unencapsulated conjugate is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, (c) humectants such as glycerol, (d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, (e) solution retarding agents such as paraffin, (f) absorption accelerators such as quaternary ammonium compounds, (g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, (h) absorbents such as kaolin and bentonite clay, and (i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets, and pills, the dosage form may also comprise buffering agents.
Disclosed nanoparticles may be formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of nanoparticle appropriate for the patient to be treated. For any nanoparticle, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. An animal model may also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic efficacy and toxicity of nanoparticles can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose is therapeutically effective in 50% of the population) and LD50 (the dose is lethal to 50% of the population). The dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50. Pharmaceutical compositions which exhibit large therapeutic indices may be useful in some embodiments. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for human use.
In an exemplary embodiment, a pharmaceutical composition is disclosed that includes a plurality of nanoparticles each comprising a therapeutic agent; and a pharmaceutically acceptable excipient.
In some embodiments, a composition suitable for freezing is comtemplated, including nanoparticles disclosed herein and a solution suitable for freezing, e.g., a sugar (e.g. sucrose) solution is added to a nanoparticle suspension. The sucrose may e.g., act as a cryoprotectant to prevent the particles from aggregating upon freezing. For example, provided herein is a nanoparticle formulation comprising a plurality of disclosed nanoparticles, sucrose and water; wherein, for example, the nanoparticles/sucrose/water is are present with about 5-10%/10-15%/80-90% (w/w/w).
Methods of Treatment
In some embodiments, therapeutic particles disclosed herein may be used to treat, alleviate, ameliorate, relieve, delay onset of, inhibit progression of, reduce severity of, and/or reduce incidence of one or more symptoms or features of a disease, disorder, and/or condition. For example, disclosed therapeutic particles, that include e.g., temsirolimus may be used to treat renal cell carcinoma. In another embodiment, disclosed therapeutic particles that include e.g. everolimus or temsirolimus may be used to treat kidney cancer, glioblastoma multiforme, mantle cell lymphoma, or dermal Kaposi's sarcoma.
Also contemplated here are methods of treating patients that have been subject to organ transplantation, by administering disclosed nanoparticles that e.g. include sirolimus. Other methods contemplated herein include methods of treating patients having tuberous sclerosis complex, and/or autism by administering an effective amount of a disclosed nanoparticle.
Methods contemplated herein include, for example, a method of preventing or deterring neointimal hyperplasia in a blood vessel of a patient, for example, a patient receiving a bare metal stent in a lesion of the blood vessel, is disclosed, comprising administering a composition comprising disclosed therapeutic particles such as those that include sirolimus or everolimus. Also contemplated herein are methods of treating or preventing restenosis (e.g. in a patient receiving a stent) comprising administering disclosed nanoparticles having e.g. sirolimus or everolimus to a patient.
Disclosed treatment methods may comprise administering a therapeutically effective amount of the disclosed therapeutic particles to a subject in need thereof, in such amounts and for such time as is necessary to achieve the desired result. In certain embodiments of the present invention a “therapeutically effective amount” is that amount effective for treating, alleviating, ameliorating, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of e.g. a cancer being treated.
Also provided herein are therapeutic protocols that include administering a therapeutically effective amount of an disclosed therapeutic particle to a healthy individual (i.e., a subject who does not display any symptoms of cancer and/or who has not been diagnosed with cancer). For example, healthy individuals may be “immunized” with an inventive targeted particle prior to development of cancer and/or onset of symptoms of cancer; at risk individuals (e.g., patients who have a family history of cancer; patients carrying one or more genetic mutations associated with development of cancer; patients having a genetic polymorphism associated with development of cancer; patients infected by a virus associated with development of cancer; patients with habits and/or lifestyles associated with development of cancer; etc.) can be treated substantially contemporaneously with (e.g., within 48 hours, within 24 hours, or within 12 hours of) the onset of symptoms of cancer. Of course individuals known to have cancer may receive inventive treatment at any time.
In other embodiments, disclosed nanoparticles may be used to inhibit the growth of cancer cells, e.g., prostate cancer cells. As used herein, the term “inhibits growth of cancer cells” or “inhibiting growth of cancer cells” refers to any slowing of the rate of cancer cell proliferation and/or migration, arrest of cancer cell proliferation and/or migration, or killing of cancer cells, such that the rate of cancer cell growth is reduced in comparison with the observed or predicted rate of growth of an untreated control cancer cell. The term “inhibits growth” can also refer to a reduction in size or disappearance of a cancer cell or tumor, as well as to a reduction in its metastatic potential. Preferably, such an inhibition at the cellular level may reduce the size, deter the growth, reduce the aggressiveness, or prevent or inhibit metastasis of a cancer in a patient. Those skilled in the art can readily determine, by any of a variety of suitable indicia, whether cancer cell growth is inhibited.
Inhibition of cancer cell growth may be evidenced, for example, by arrest of cancer cells in a particular phase of the cell cycle, e.g., arrest at the G2/M phase of the cell cycle. Inhibition of cancer cell growth can also be evidenced by direct or indirect measurement of cancer cell or tumor size. In human cancer patients, such measurements generally are made using well known imaging methods such as magnetic resonance imaging, computerized axial tomography and X-rays. Cancer cell growth can also be determined indirectly, such as by determining the levels of circulating carcinoembryonic antigen, prostate specific antigen or other cancer-specific antigens that are correlated with cancer cell growth. Inhibition of cancer growth is also generally correlated with prolonged survival and/or increased health and well-being of the subject.
EXAMPLES
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention in any way.
Example 1 Preparation of PLA-PEG
The synthesis is accomplished by ring opening polymerization of d,l-lactide with α-hydroxy-ω-methoxypoly(ethylene glycol) as the macro-initiator, and performed at an elevated temperature using Tin (II) 2-Ethyl hexanoate as a catalyst, as shown below (PEG Mn≈5,000 Da; PLA Mn≈16,000 Da; PEGPLA Mn≈21,000 Da)
Figure US08613951-20131224-C00003
The polymer is purified by dissolving the polymer in dichloromethane, and precipitating it in a mixture of hexane and diethyl ether. The polymer recovered from this step shall be dried in an oven.
Example 2 Nanoparticle Preparation—Emulsion Process
An organic phase is formed composed of a mixture of sirolimus and polymer (homopolymer, co-polymer, and co-polymer with ligand). The organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent. In order to achieve high drug loading, about 30% solids in the organic phase is used.
The primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer. The rotor/stator yielded a homogeneous milky solution, while the stir bar produced a visibly larger coarse emulsion. It was observed that the stir bar method resulted in significant oil phase droplets adhering to the side of the feed vessel, suggesting that while the coarse emulsion size is not a process parameter critical to quality, it should be made suitably fine in order to prevent yield loss or phase separation. Therefore the rotor stator is used as the standard method of coarse emulsion formation, although a high speed mixer may be suitable at a larger scale.
The primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer. The size of the coarse emulsion does not significantly affect the particle size after successive passes (1-3) through the homogenizer. M-110-EH (FIG. 3).
Homogenizer feed pressure was found to have a significant impact on resultant particle size. On both the pneumatic and electric M-110EH homogenizers, it was found that reducing the feed pressure also reduced the particle size (FIG. 4). Therefore the standard operating pressure used for the M-110EH is 4000-5000 psi per interaction chamber, which is the minimum processing pressure on the unit. The M-110EH also has the option of one or two interaction chambers. It comes standard with a restrictive Y-chamber, in series with a less restrictive 200 μm Z-chamber. It was found that the particle size was actually reduced when the Y-chamber was removed and replaced with a blank chamber. Furthermore, removing the Y-chamber significantly increases the flow rate of emulsion during processing.
After 2-3 passes the particle size was not significantly reduced, and successive passes can even cause a particle size increase. The results are summarized in FIG. 5.
The effect of scale on particle size showed surprising scale dependence. The trend shows that in the 2-10 g batch size range, larger batches produce smaller particles. It has been demonstrated that this scale dependence is eliminated when considering greater than 10 g scale batches. The amount of solids used in the oil phase was about 30%. FIG. 6 depicts the effect of solids concentration on particle size.
Table A summarizes the emulsification process parameters.
TABLE A
Parameter Value Observation
Coarse emulsion Rotor stator Coarse emulsion size does not affect final particle size, but large
formation homogenizer coarse emulsion can cause increased oil phase retention in feed vessel
Homogenizer feed 4000-5000 psi per Lower pressure reduces particle size
pressure chamber
Interaction
2 × 200 μm Z- 200 μm Z-chamber yields the smallest particle size, and allows for
chamber(s) chamber highest homogenizer throughput
Number of 2-3 passes Studies have shown that the particle size is not significantly reduced
homogenizer passes after 2 discreet passes, and size can even increase with successive
passes
Water phase 0.1% [Sodium cholate] can effectively alter particle size; value is optimized
[sodium cholate] for given process and formulation
W:O ratio 5:1 Lowest ratio without significant particle size increase is ~5:1
[Solids] in oil phase 30% Increased process efficiency, increased drug encapsulation, workable
viscosity
The fine emulsion is then quenched by addition to deionized water at a given temperature under mixing. In the quench unit operation, the emulsion is added to a cold aqueous quench under agitation. This serves to extract a significant portion of the oil phase solvents, effectively hardening the nanoparticles for downstream filtration. Chilling the quench significantly improved drug encapsulation. The quench:emulsion ratio is approximately 5:1.
A solution of 35% (wt %) of Tween 80 is added to the quench to achieve approximately 2% Tween 80 overall After the emulsion is quenched a solution of Tween-80 is added which acts as a drug solubilizer, allowing for effective removal of unencapsulated drug during filtration. Table B indicates each of the quench process parameters.
TABLE B
Summary quench process parameters.
Parameter Value Observation
Initial quench <5° C. Low temperature yields higher
temperature drug encapsulation
[Tween-80] solution 35% Highest concentration that
can be prepared and readily
disperses in quench
Tween-80:drug ratio 25:1 Minimum amount of Tween-80
required to effectively remove
unencapsulated drug
Q:E ratio  5:1 Minimum Q:E ratio while
retaining high drug
encapsulation
Quench ≦5° C. Temperature which prevents
hold/processing temp (with current significant drug leaching during
5:1 Q:E ratio, quench hold time and initial
25:1 Tween- concentration step
80:drug ratio)
The temperature must remain cold enough with a dilute enough suspension (low enough concentration of solvents) to remain below the Tg of the particles. If the Q:E ratio is not high enough, then the higher concentration of solvent plasticizes the particles and allows for drug leakage. Conversely, colder temperatures allow for high drug encapsulation at low Q:E ratios (to ˜3:1), making it possible to run the process more efficiently.
The nanoparticles are then isolated through a tangential flow filtration process to concentrate the nanoparticle suspension and buffer exchange the solvents, free drug, and drug solubilizer from the quench solution into water. A regenerated cellulose membrane is used with a molecular weight cutoffs (MWCO) of 300.
A constant volume diafiltration (DF) is performed to remove the quench solvents, free drug and Tween-80. To perform a constant-volume DF, buffer is added to the retentate vessel at the same rate the filtrate is removed. The process parameters for the TFF operations are summarized in Table C. Crossflow rate refers to the rate of the solution flow through the feed channels and across the membrane. This flow provides the force to sweep away molecules that can foul the membrane and restrict filtrate flow. The transmembrane pressure is the force that drives the permeable molecules through the membrane.
TABLE C
TFF Parameters
Optimized
Parameter Value Effect
Membrane Regenerated No difference in performance between RC and PES, but
Material cellulose - solvent compatibility is superior for RC.
Coarse Screen
Membrane
Molecular Weight 300 kDa No difference in NP characteristics (i.e. residual
Cut off tween)Increase in flux rates is seen with 500 kDa
membrane but 500 kDa is not available in RC
Crossflow Rate 11 L/min/m2 Higher crossflow rate led to higher flux
Transmembrane
20 psid Open channel membranes have maximum flux rates
Pressure between 10 and 30 psid. Coarse channel membranes have
maximum flux rates with min TMP (~20 psid).
Concentration of 30 mg/ml Diafiltration is most efficient at [NP] ~50 mg/ml with
Nanoparticle open channel TFF membranes based on flux rates and
Suspension for throughput. With coarse channel membranes the flux rate
Diafiltration is optimized at ~30 mg/ml in the starting buffer.
Number of ≧15 (based on About 15 diavolumes are needed to effectively remove
Diavolumes flux increase) tween-80. End point of diafiltration is determined by in-
process control (flux increase plateau).
Membrane Area ~1 m2/kg Membranes sized based on anticipated flux rates and
volumes required.
The filtered nanoparticle slurry is then thermal cycled to an elevated temperature during workup. A small portion (typically 5-10%) of the encapsulated drug is released from the nanoparticles very quickly after its first exposure to 25° C. Because of this phenomenon, batches that are held cold during the entire workup are susceptible to free drug or drug crystals forming during delivery or any portion of unfrozen storage. By exposing the nanoparticle slurry to elevated temperature during workup, this ‘loosely encapsulated’ drug can be removed and improve the product stability at the expense of a small drop in drug loading. 5 diavolumes is used as the amount for cold processing prior to the 25° C. treatment.
After the filtration process the nanoparticle suspension is passed through a sterilizing grade filter (0.2 μm absolute). Pre-filters are used to protect the sterilizing grade filter in order to use a reasonable filtration area/time for the process. Values are as summarized in Table D.
TABLE D
Parameter O Value Effect
Nanoparticle    50 mg/ml Yield losses are higher at higher [NP],
Suspension but the ability to filter at 50 mg/ml
Concentration obviates the need to aseptically
concentrate after filtration
Filtration flow ~1.3 L/min/m2 Filterability decreases as flow rate
rate increases
The filtration train is Ertel Alsop Micromedia XL depth filter M953P membrane (0.2 μm Nominal); Pall SUPRAcap with Seitz EKSP depth filter media (0.1-0.3 μm Nominal); Pall Life Sciences Supor EKV 0.65/0.2 micron sterilizing grade PES filter.
0.2 m2 of filtration surface area per kg of nanoparticles for depth filters and 1.3 m2 of filtration surface area per kg of nanoparticles for the sterilizing grade filters can be used.
Example 3 Cryoprotectant
Freezing a suspension of nanoemulsion nanoparticles in deionized water alone results in particle aggregation. This is believed to be due to crystallization and entanglement of PEG chains on the nanoparticle surfaces. Sugar-based excipients (sucrose, trehalose, or mannitol) can act to cryoprotect these nanoparticles under freeze/thaw conditions, with a concentrations as low as 1 wt % for dilute (˜10 mg/ml) nanoparticle suspensions. One formulation includes 10 wt % sucrose, which contains excess sucrose to what is required and is the same osmolality as physiological saline.
Table E shows that 16/5 PLA-PEG co-polymer is less susceptible to freeze-thaw aggregation.
TABLE E
Post-
Original F/T
Median Median Post-F/T Post-F/T
PSD/ PS Poly- Baseline
Description PD (nm) dispersity Index
1:1 45/5 and PLA 143.4, 0.124 358.9 0.358 0.0/23.16%
(baseline)
16/5 PLA-PEG and 186.7, 0.080 189.5 0.126 9.7/91.57%
PLA (1:1)
2:1:1 16/5:PLA:cetyl 174.1, 0.084 232.7 0.146 0.0/61.19%
2:1:1 45/5:PLA:cetyl 111.0, 0.182 0 0 0.0/1.55%
16/5 PLA-PEG alone 218.8, 0.098 226.9 0.03 7.3/60.56%
16/5 PLA-PEG and 222.2, 0.126 230.7 0.065 4.1/35.36%
PLA (3:1)
45/5 PLGA-PEG and 162.7, 0.099 178.6 0.091 7.7/95.41%
PLA (3:1)
2:1:1 45/5 PLA- 115.9, 0.154 734.6 0.392 0.0/13.27%
PEG:PLA:cetyl
Example 4 In Vitro Release
An in vitro release method is used to determine the initial burst phase release from nanoparticles at both ambient and 37° C. conditions. In order to maintain sink conditions and prevent nanoparticles from entering the release samples, a dialysis system was designed. After obtaining an ultracentrifuge capable of pelleting 100 nm particles, the dialysis membranes were eliminated and centrifugation was used to separate released drug from encapsulated drug.
The dialysis system is as follows: 3 mL slurry of sirolimus nanoparticles (approx 250 μg/mL sirolimus PLGA/PLA nanoparticles, corresponding to 2.5 mg/mL solid concentration) in DI-water is placed into the inner tube of a 300 kDa MWCO dialyzer by pipetting. The nanoparticle is suspension in this media. The dialyzer is placed into a glass bottles containing 130 ml release media (2.5% hydroxyl beta cyclodextrin in PBS), which is continually stirred at 150 rpm using a shaker to prevent the formation of an unstirred water layer at the membrane/outer solution interface. At pre-determined time points, aliquot of samples (1 mL) were withdrawn from the outer solution (dialysate) and analyzed for sirolimus concentration by HPLC.
The centrifugal system is run using similar conditions at lower suspension volumes without dialysis bags. Samples are centrifuged at 60,000 g for 30 minutes and the supernatant is assayed for sirolimus content to measured released sirolimus.
Example 5 Particle Size Analysis
Particle size is analyzed by two techniques—dynamic light scattering (DLS) and laser diffraction. DLS is performed using a Brookhaven ZetaPals instrument at 25° C. in dilute aqueous suspension using a 660 nm laser scattered at 90° and analyzed using the Cumulants and NNLS methods (TP008). Laser diffraction is performed with a Horiba LS950 instrument in dilute aqueous suspension using both a HeNe laser at 633 nm and an LED at 405 nm, scattered at 90° and analyzed using the Mie optical model (TP009). The output from the DLS is associated with the hydrodynamic radius of the particles, which includes the PEG ‘corona’, while the laser diffraction instrument is more closely associated with the geometric size of the PLA particle ‘core’.
Example 6
Nanoparticle batches were prepared using the general procedure of Example 2, with 80% (w/w) Polymer-PEG or Polymer-PEG with homopolymer PLA at 40% (w/w) each, with a batch of % total solids of 5%, 15% and 30%. Solvents used were: 21% benzyl alcohol and 79% ethyl acetate (w/w). For each 2 gram batch size, 400 mg of drug was used and 1.6 g of 16-5 Polymer-PEG or 0.8 g of 16-5 Polymer-PEG+0.8 g of 10 kDa PLA (homopolymer) was used. The diblock polymer 16-5 PLA-PEG or PLGA-PEG (50:50 L:G) was used, and if used, the homopolymer: PLA with a Mn=6.5 kDa, Mw=10 kDa, and Mw/Mn=1.55.
The organic phase (drug and polymer) is prepared in 2 g batches: To 20 mL scintillation vial add drug and polymer(s). The mass of solvents needed at % solids concentration is shown below:
i. 5% solids: 7.98 g benzyl alcohol+30.02 g ethyl acetate
ii. 15% solids: 2.38 g benzyl alcohol+8.95 g ethyl acetate
iii. 30% solids: 0.98 g benzyl alcohol+3.69 g ethyl acetate
An aqueous solution is prepared with 0.5% sodium cholate, 2% benzyl alcohol, and 4% ethyl acetate in water. To a 2 L bottle add 7.5 g sodium cholate, 1402.5 g of DI water, 30 g of benzyl alcohol and 60 g of ethyl acetate, and mix on stir plate until dissolved.
For the formation of emulsion, a ratio of aqueous phase to oil phase of 5:1 is used. The organic phase is poured into the aqueous solution and homogenized using IKA for 10 seconds at room temperature to form course emulsion. The solution is fed through the homogenizer (110S) at 9 Kpsi (45 psi on gauge) for 2 discreet passes to form nanoemulsion.
The emulsion is poured into quench (D.I. water) at <5 C while stirring on stir plate. Ratio of quench to emulsion is 8:1. 35% (w/w) Tween 80 is added in water to quench at ratio of 25:1 Tween 80 to drug. The nanoparticles are concentrated through TFF and the quench is concentrated on TFF with 500 kDa Pall cassette (2 membrane) to ˜100 mL. Diafiltering is used using ˜20 diavolumes (2 liter) of cold DI water, and the volume is brought down to minimal volume then collect final slurry, ˜100 mL. The solids concentration of unfiltered final slurry is determined by the using tared 20 mL scintillation vial and adding 4 mL final slurry and dry under vacuum on lyo/oven and the weight of nanoparticles in the 4 mL of slurry dried down is determined. Concentrated sucrose (0.666 g/g) is added to final slurry sample to attain 10% sucrose.
Solids concentration of 0.45 um filtered final slurry was determined by filtering about 5 mL of final slurry sample before addition of sucrose through 0.45 μm syringe filter; to tared 20 mL scintillation vial add 4 mL of filtered sample and dry under vacuum on lyo/oven.
The remaining sample of unfiltered final slurry was frozen with sucrose. Rapamycin (sirolimus) formulations:
Drug Release of Drug (t = hr)
Name Polymer Size (nm) Loading T = 0 T = 2 T = 4 T = 24
 5% Solid 16/5 PLA/PEG 123.1 3.61% ND ND ND ND
16/5 PLA/PEG + PLA 119.7 4.49% ND ND ND ND
15% Solid 16/5 PLA/PEG 82.1 4.40% ND ND ND ND
16/5 PLA/PEG + PLA 120.6 11.51%  ND ND ND ND
23% Solid 16/5 PLA/PEG 88.1 7.40% ND ND ND ND
16/5 PLA/PEG + PLA 118.3  7.8% ND ND ND ND
30% Solid 16/5 PLA/PEG 88.5 10.26%  8.5 17.3 22.4 64.2
16/5 PLA/PEG + PLA 118.3 10.18%  9.3 30.4 44.7 98.2
The effect of solid contents and the inclusions of poly(lactic) acid homopolymer is shown in FIG. 7.
In-vitro release experiments are studied by dispersing nanoparticles in PBS containing 10% (w/w) of Tween 20 (T20) at 37° C. T20 was used to increase the solubility of rapamycin in PBS to levels well detectable by HPLC as well as maintaining the sink condition. 3 mL of drug-loaded nanoparticles were redispersed in 130 mL of release medium in a jar at a known concentration (approximately 250 μg/ml). These volumes were chosen to ensure that the maximum concentration of the drug in the release medium would always be less than 10% of the maximum solubility, i.e., sink conditions. The media and nanoparticle suspension is stirred at 150 rpm. At pre-determined time points, 4 ml of aliquots were centrifuged at 50,000 rpm (236,000 g) for 1 hr to separate the nanoparticles from the elution media. The elution media is injected in to a HPLC to determine drug released from the nanoparticles. The release of rapamycin showed slow and sustained release, as shown in FIG. 8.
Example 7
Nanoparticles were prepared as in Example 2 and 6, except temsirolimus was used with 30% solid content in the organic phase before emulsion:
Size Drug Release of Drug (t = hr)
Name Lot # Polymer (nm) Loading T = 0 T = 2 T = 4 T = 24
30% 45-48-1 16/5 PLA/PEG 97.5 9.9% 11.5 15.6 17.9 40.9
Solid 45-48-2 16/5 PLA/PEG + 112.8 14.2% 9.8 22.3 29.9 88.0
PLA
45-100-1 16/5 PLGA/PEG + 150.3 4.6 ND ND ND ND
PLA
50-52-6 16/5 PLGA/PEG + ND 6.9 10.6 35.7 45.8 87.0
PLA
FIG. 9 depicts the weight % of temsirolimus and FIG. 10 depicts the nanoparticle for the different polymeric nanoparticles having temsirolimus. The results of an in-vitro release experiment as in Example 6 shows the slow and sustained release of temsirolimus showed slow and sustained release, as shown in FIG. 11.
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
INCORPORATION BY REFERENCE
The entire contents of all patents, published patent applications, websites, and other references cited herein are hereby expressly incorporated herein in their entireties by reference.

Claims (24)

The invention claimed is:
1. A therapeutic nanoparticle comprising:
about 5 to about 15 weight percent of a mTOR inhibitor; and
about 50 to about 99 weight percent biocompatible polymer, wherein the biocompatible polymer is a diblock poly(lactic) acid-poly(ethylene)glycol copolymer, wherein said diblock poly(lactic) acid-poly(ethylene)glycol copolymer comprises poly(lactic acid) having a number average molecular weight of about 15 to 20 kDa and poly(ethylene)glycol having a number average molecular weight of about 4 to about 6 kDa, wherein the diameter of the therapeutic nanoparticle is about 70 to about 130 nm; and
wherein the particle releases less than about 10% of the mTOR inhibitor over about one minute when placed in a phosphate buffer solution at room temperature.
2. The therapeutic nanoparticle of claim 1 wherein said mTOR inhibitor is sirolimus, temsirolimus, or everolimus or a pharmaceutically acceptable salt thereof.
3. The therapeutic nanoparticle of claim 1, wherein the diameter is about 80 to about 130 nm.
4. The therapeutic nanoparticle of claim 1, wherein said diblock poly(lactic) acid-poly(ethylene)glycol copolymer comprises poly(lactic acid) having a number average molecular weight of about 16 kDa and poly(ethylene)glycol having a number average molecular weight of about 5 kDa.
5. A therapeutic nanoparticle comprising:
about 10 to about 20 weight percent of a therapeutic agent selected from the group consisting of sirolimus, temsirolimus, or everolimus; and
a diblock poly(lactic) acid-poly(ethylene)glycol copolymer,
wherein said diblock poly(lactic) acid-poly(ethylene)glycol copolymer comprises poly(lactic acid) having a number average molecular weight of about 15 to 20 kDa and poly(ethylene)glycol having a number average molecular weight of about 4 to about 6 kDa, wherein the diameter of the therapeutic nanoparticle is about 70 to about 130 nm; and wherein the particle releases about 0.1 to about 20% therapeutic agent over about one hour when placed in an aqueous solution at 25° C.
6. The therapeutic nanoparticle of claim 5, wherein the particle substantially immediately releases less than about 4% of the therapeutic agent when placed in a phosphate buffer solution at room temperature.
7. The therapeutic nanoparticle of claim 5, wherein the particle substantially immediately releases less than about 2% of the therapeutic agent when placed in a phosphate buffer solution at room temperature.
8. The therapeutic nanoparticle of claim 1, further comprising cetyl alcohol.
9. A method of treating dermal Kaposi's sarcoma, lymphomas, glioblastoma multiforme, mantle cell lymphoma, renal cell carcinoma, comprising administering to a patient in need thereof an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
10. A method of ameliorating organ rejection after transplant in a patient in need thereof, comprising administering to the patient an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
11. A method of treating, preventing or deterring restenosis in a patient in need thereof, comprising administering to the patient an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
12. A method of treating or deterring neointimal hyperplasia in a patient in need thereof, comprising administering to the patient an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
13. A method of treating autosomal dominant polycystic kidney disease, comprising administering to the patient an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
14. A method of treating tuberous sclerosis complex, comprising administering to the patient an effective amount of a composition comprising the therapeutic nanoparticle of claim 1.
15. The therapeutic nanoparticle of claim 1, wherein the diameter of the nanoparticle is about 70 to about 120 nm.
16. The therapeutic nanoparticle of claim 1, wherein the nanoparticle substantially immediately releases less than about 10% of the mTOR inhibitor over 30 minutes when placed in a phosphate buffer solution at 37° C.
17. The therapeutic nanoparticle of claim 1, wherein the mTOR inhibitor is everolimus.
18. The therapeutic nanoparticle of claim 5, wherein the therapeutic agent is everolimus.
19. A pharmaceutical composition comprising:
a plurality of nanoparticles each comprising about 4 to about 25 weight percent of a mTOR inhibitor, about 50 to about 99 weight percent of a diblock poly(lactic) acid-poly(ethylene)glycol copolymer comprising poly(lactic acid) having a number average molecular weight of about 15 to 20 kDa and poly(ethylene)glycol having a number average molecular weight of about 4 to about 6 kDa, and having a diameter of about 70 to about 140 nm; and
a saccharide;
wherein said nanoparticles are stable for at least 3 days when held at 25° C.
20. The pharmaceutical composition of claim 19, wherein said nanoparticles substantially retains the mTOR inhibitor for at least 5 days at 25° C.
21. The pharmaceutical composition of claim 19, comprising sucrose.
22. The pharmaceutical composition of claim 19, wherein said nanoparticles release less than about 10% mTOR inhibitor when placed in a phosphate buffer solution at 25° C. for 30 minutes.
23. The pharmaceutical composition of claim 19, wherein the mTOR inhibitor is everolimus.
24. The pharmaceutical composition of claim 19, wherein the poly(lactic) acid of the copolymer has a number average molecular weight of about 16 kDa and the poly(ethylene)glycol has a number average molecular weight of about 5 kDa.
US12/485,462 2008-06-16 2009-06-16 Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same Expired - Fee Related US8613951B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/485,462 US8613951B2 (en) 2008-06-16 2009-06-16 Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US13/910,328 US8623417B1 (en) 2008-06-16 2013-06-05 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US14/091,343 US9579284B2 (en) 2008-06-16 2013-11-27 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
US6176008P 2008-06-16 2008-06-16
US6169708P 2008-06-16 2008-06-16
US6170408P 2008-06-16 2008-06-16
US8815908P 2008-08-12 2008-08-12
US10591608P 2008-10-16 2008-10-16
US10677708P 2008-10-20 2008-10-20
US16951409P 2009-04-15 2009-04-15
US16951909P 2009-04-15 2009-04-15
US16954109P 2009-04-15 2009-04-15
US17379009P 2009-04-29 2009-04-29
US17521909P 2009-05-04 2009-05-04
US17520909P 2009-05-04 2009-05-04
US17522609P 2009-05-04 2009-05-04
US12/485,462 US8613951B2 (en) 2008-06-16 2009-06-16 Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/910,328 Continuation US8623417B1 (en) 2008-06-16 2013-06-05 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same

Publications (2)

Publication Number Publication Date
US20100069426A1 US20100069426A1 (en) 2010-03-18
US8613951B2 true US8613951B2 (en) 2013-12-24

Family

ID=41507663

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/485,462 Expired - Fee Related US8613951B2 (en) 2008-06-16 2009-06-16 Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US13/910,328 Expired - Fee Related US8623417B1 (en) 2008-06-16 2013-06-05 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US14/091,343 Active 2029-08-31 US9579284B2 (en) 2008-06-16 2013-11-27 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/910,328 Expired - Fee Related US8623417B1 (en) 2008-06-16 2013-06-05 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US14/091,343 Active 2029-08-31 US9579284B2 (en) 2008-06-16 2013-11-27 Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same

Country Status (2)

Country Link
US (3) US8613951B2 (en)
WO (1) WO2010005726A2 (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120276162A1 (en) * 2008-06-16 2012-11-01 Zale Stephen E Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20140186453A1 (en) * 2008-06-16 2014-07-03 Bind Therapeutics, Inc. Therapeutic Polymeric Nanoparticles with mTOR Inhibitors and Methods of Making and Using Same
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8905997B2 (en) 2008-12-12 2014-12-09 Bind Therapeutics, Inc. Therapeutic particles suitable for parenteral administration and methods of making and using same
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8987237B2 (en) 2011-11-23 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US20160045608A1 (en) * 2008-12-15 2016-02-18 Bind Therapeutics, Inc. Long circulating nanoparticles for sustained release of therapeutic agents
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9351933B2 (en) 2008-06-16 2016-05-31 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US9877923B2 (en) 2012-09-17 2018-01-30 Pfizer Inc. Process for preparing therapeutic nanoparticles
US9895378B2 (en) 2014-03-14 2018-02-20 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10266485B2 (en) 2015-09-17 2019-04-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11260027B2 (en) 2015-07-29 2022-03-01 Musc Foundation For Research Development Donor organ pre-treatment formulation
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations

Families Citing this family (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492400B2 (en) * 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
WO2007070682A2 (en) 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
ES2776100T3 (en) 2006-03-31 2020-07-29 Massachusetts Inst Technology System for targeted delivery of therapeutic agents
US20110052697A1 (en) * 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
US9381477B2 (en) * 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US20100144845A1 (en) * 2006-08-04 2010-06-10 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20100303723A1 (en) * 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
EP2134830A2 (en) 2007-02-09 2009-12-23 Massachusetts Institute of Technology Oscillating cell culture bioreactor
WO2008121949A1 (en) 2007-03-30 2008-10-09 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
JP2010523595A (en) 2007-04-04 2010-07-15 マサチューセッツ インスティテュート オブ テクノロジー Poly (amino acid) targeting part
JP2011500569A (en) 2007-10-12 2011-01-06 マサチューセッツ インスティテュート オブ テクノロジー Vaccine nanotechnology
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
KR20120022984A (en) * 2009-04-21 2012-03-12 셀렉타 바이오사이언시즈, 인크. Immunonanotherapeutics providing a th1-biased response
BRPI1012036A2 (en) 2009-05-27 2017-10-10 Selecta Biosciences Inc nanocarriers that have components with different release rates
EA036522B1 (en) 2009-12-11 2020-11-19 Пфайзер Инк. Pharmaceutical composition suitable for lyophilization comprising a plurality of therapeutic particles
EA201290499A1 (en) 2009-12-15 2013-01-30 Байнд Байосайенсиз, Инк. COMPOSITIONS OF THERAPEUTIC POLYMER NANOPARTICLES WITH HIGH GLASSING TEMPERATURE AND HIGH-MOLECULAR CO-POLYMERS
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
WO2011119262A1 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Methods and systems for generating nanoparticles
JP6367554B2 (en) 2010-05-26 2018-08-01 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Dosage selection of adjuvanted synthetic nanocarriers
WO2012068531A2 (en) 2010-11-18 2012-05-24 The General Hospital Corporation Novel compositions and uses of anti-hypertension agents for cancer therapy
US8546521B2 (en) 2011-01-28 2013-10-01 Cerulean Pharma Inc. Method for fabricating nanoparticles
CA2834571A1 (en) 2011-04-29 2012-11-01 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory b cells
CN103702687A (en) 2011-07-29 2014-04-02 西莱克塔生物科技公司 Synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte (CTL) immune responses
CN108949772A (en) 2012-04-02 2018-12-07 现代泰克斯公司 For generating the modification polynucleotides of biological agent relevant to human diseases and protein
CN104736181A (en) 2012-08-10 2015-06-24 北德克萨斯大学健康科学中心 Drug delivery vehicle comprising conjugates between targeting polyamino acids and fatty acids
WO2014179762A1 (en) 2013-05-03 2014-11-06 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers and therapeutic macromolecules for reduced or enhanced pharmacodynamic effects
HUE056760T2 (en) 2013-07-11 2022-03-28 Modernatx Inc Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
SI3311845T1 (en) 2013-09-16 2020-06-30 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
WO2015051349A1 (en) * 2013-10-04 2015-04-09 Sorrento Therapeutics, Inc. Treating metastatic cancer with micellular paclitaxel
SI3071696T1 (en) 2013-11-22 2019-11-29 Mina Therapeutics Ltd C/ebp alpha short activating rna compositions and methods of use
CA2953341C (en) 2014-06-25 2023-01-24 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
JP2017524357A (en) 2014-07-16 2017-08-31 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chimeric polynucleotide
EP3171895A1 (en) 2014-07-23 2017-05-31 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
MX2017002935A (en) 2014-09-07 2017-05-30 Selecta Biosciences Inc Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses.
US10583091B2 (en) 2014-10-23 2020-03-10 The Brigham And Women's Hospital, Inc. Amphiphile-polymer particles
EP3906918B1 (en) * 2014-11-05 2024-01-03 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
IL283545B2 (en) 2015-06-29 2023-09-01 Acuitas Therapeutics Inc Lipids and lipid nanoparticle formulations for delivery of nucleic acids
SI3718565T1 (en) 2015-10-22 2022-08-31 Modernatx, Inc. Respiratory virus vaccines
AU2016341311B2 (en) 2015-10-22 2023-11-16 Modernatx, Inc. Respiratory syncytial virus vaccine
AU2016342371B2 (en) 2015-10-22 2023-05-11 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
JP6921833B2 (en) 2015-10-22 2021-08-18 モデルナティーエックス, インコーポレイテッド Human cytomegalovirus vaccine
CA3003103A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Broad spectrum influenza virus vaccine
MX2018004918A (en) 2015-10-22 2019-04-01 Modernatx Inc Herpes simplex virus vaccine.
AU2016342027B2 (en) 2015-10-23 2021-05-13 Navitor Pharmaceuticals, Inc. Modulators of Sestrin-GATOR2 interaction and uses thereof
RS63986B1 (en) 2015-10-28 2023-03-31 Acuitas Therapeutics Inc Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
SI3394093T1 (en) 2015-12-23 2022-05-31 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
MA43587A (en) 2016-01-10 2018-11-14 Modernatx Inc THERAPEUTIC RNA CODING FOR ANTI-CTLA-4 ANTIBODIES
US11471515B2 (en) 2016-11-09 2022-10-18 The Brigham And Women's Hospital, Inc. Restoration of tumor suppression using MRNA-based delivery system
CA3055936A1 (en) 2017-03-11 2018-09-20 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with anti-inflammatories and synthetic nanocarriers comprising an immunosuppressant
WO2018200625A1 (en) 2017-04-26 2018-11-01 Navitor Pharmaceuticals, Inc. Modulators of sestrin-gator2 interaction and uses thereof
CN110799492B (en) 2017-04-28 2023-06-27 爱康泰生治疗公司 Novel carbonyl lipid and lipid nanoparticle formulations for delivery of nucleic acids
US11485972B2 (en) 2017-05-18 2022-11-01 Modernatx, Inc. Modified messenger RNA comprising functional RNA elements
AU2018270111B2 (en) 2017-05-18 2022-07-14 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (IL12) polypeptides and uses thereof
EP3638292A1 (en) 2017-06-14 2020-04-22 ModernaTX, Inc. Polynucleotides encoding coagulation factor viii
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
EP4219715A3 (en) 2017-09-08 2023-09-06 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
JP7424976B2 (en) 2017-11-22 2024-01-30 モダーナティエックス・インコーポレイテッド Polynucleotide encoding propionyl-CoA carboxylase alpha and beta subunits for the treatment of propionic acidemia
CA3079428A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
MA51523A (en) 2018-01-05 2020-11-11 Modernatx Inc POLYNUCLEOTIDES CODING ANTI-BODY ANTI-CHIKUNGUNYA VIRUS
EP3773477A1 (en) 2018-04-03 2021-02-17 Vaxess Technologies, Inc. Microneedle comprising silk fibroin applied to a dissolvable base
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
EP3775211B1 (en) 2018-04-12 2023-04-05 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
MA52709A (en) 2018-05-23 2021-03-31 Modernatx Inc DNA ADMINISTRATION
US20220184185A1 (en) 2018-07-25 2022-06-16 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
EP3846776A1 (en) 2018-09-02 2021-07-14 ModernaTX, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
CN113164561A (en) 2018-09-13 2021-07-23 摩登纳特斯有限公司 Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
US20220243182A1 (en) 2018-09-13 2022-08-04 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
EP3850102A1 (en) 2018-09-14 2021-07-21 ModernaTX, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
AU2019364542A1 (en) 2018-10-24 2021-06-10 Navitor Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
MX2021008358A (en) 2019-01-11 2021-09-30 Acuitas Therapeutics Inc Lipids for lipid nanoparticle delivery of active agents.
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
EP3965797A1 (en) 2019-05-08 2022-03-16 AstraZeneca AB Compositions for skin and wounds and methods of use thereof
EP3986480A1 (en) 2019-06-24 2022-04-27 ModernaTX, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
US20220378754A1 (en) * 2019-09-11 2022-12-01 Yale University Compositions and methods for treating slow-flow vascular malformations
IL292612A (en) 2019-11-01 2022-07-01 Navitor Pharm Inc Methods of treatment using an mtorc1 modulator
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
CA3181680A1 (en) 2020-06-12 2021-12-16 University Of Rochester Encoding and expression of ace-trnas
AU2021377895A1 (en) 2020-11-13 2023-06-15 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
CA3214137A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022240806A1 (en) 2021-05-11 2022-11-17 Modernatx, Inc. Non-viral delivery of dna for prolonged polypeptide expression in vivo
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023150753A1 (en) 2022-02-07 2023-08-10 University Of Rochester Optimized sequences for enhanced trna expression or/and nonsense mutation suppression
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
WO2023250117A2 (en) 2022-06-24 2023-12-28 Vaxess Technologies, Inc. Applicator for medicament patch
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression

Citations (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5302401A (en) 1992-12-09 1994-04-12 Sterling Winthrop Inc. Method to reduce particle size growth during lyophilization
US5543158A (en) * 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5563122A (en) 1991-12-09 1996-10-08 Asahi Kasei Kogyo Kabushiki Kaisha Stabilized parathyroid hormone composition
US5578325A (en) 1993-07-23 1996-11-26 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
EP0805678A1 (en) 1995-01-05 1997-11-12 THE BOARD OF REGENTS acting for and on behalf of THE UNIVERSITY OF MICHIGAN Surface-modified nanoparticles and method of making and using same
US5766635A (en) 1991-06-28 1998-06-16 Rhone-Poulenc Rorer S.A. Process for preparing nanoparticles
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US6136846A (en) 1999-10-25 2000-10-24 Supergen, Inc. Formulation for paclitaxel
US6139870A (en) 1995-12-19 2000-10-31 Aventis Pharma Sa Stabilized nanoparticles which are filterable under sterile conditions
US6201072B1 (en) 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6254890B1 (en) 1997-12-12 2001-07-03 Massachusetts Institute Of Technology Sub-100nm biodegradable polymer spheres capable of transporting and releasing nucleic acids
US6265609B1 (en) 1998-07-06 2001-07-24 Guilford Pharmaceuticals Inc. Thio-substituted pentanedioic acid derivatives
US6346274B1 (en) 1995-03-10 2002-02-12 Roche Diagnostics Gmbh Polypeptide-containing pharmaceutical forms of administration in the form of microparticles and method for the preparation thereof
US20020045582A1 (en) 1997-12-31 2002-04-18 Alexey L. Margolin Stabilized protein crystals formulations containing them and methods of making them
US6395718B1 (en) 1998-07-06 2002-05-28 Guilford Pharmaceuticals Inc. Pharmaceutical compositions and methods of inhibiting angiogenesis using naaladase inhibitors
KR20020041712A (en) 2000-11-28 2002-06-03 윤덕용 Process for Preparing Sustained Release Form of Micelle Employing Conjugate of Anticancer Drug and Biodegradable Polymer
US20020119916A1 (en) 2000-12-22 2002-08-29 Hassan Emadeldin M. Elemental nanoparticles of substantially water insoluble materials
US6528499B1 (en) 2000-04-27 2003-03-04 Georgetown University Ligands for metabotropic glutamate receptors and inhibitors of NAALADase
US20030068377A1 (en) 2001-10-03 2003-04-10 Macromed, Incorporated PLA/PLGA oligomers combined with block copolymers for enhancing solubility of a drug in water
WO2003055469A1 (en) 2001-12-21 2003-07-10 Celator Technologies Inc. Improved polymer-lipid delivery vehicles
US20030143184A1 (en) 2000-05-17 2003-07-31 Min-Hyo Seo Stable polymeric micelle-type drug composition and method for the preparation thereof
US20030232887A1 (en) 2002-04-10 2003-12-18 Johnson Douglas Giles Preparation and use of a stable formulation of allosteric effector compounds
US20030235619A1 (en) 2001-12-21 2003-12-25 Christine Allen Polymer-lipid delivery vehicles
US20040054190A1 (en) 2002-01-10 2004-03-18 Johns Hopkins University Imaging agents and methods of imaging NAALADase or PSMA
US20040071768A1 (en) 1999-04-01 2004-04-15 Inex Pharmaceuticals Corporation Compositions and methods for treating cancer
US20040081688A1 (en) 2000-12-27 2004-04-29 Del Curto Maria Dorly Amphiphilic lipid nanoparticles for peptide and/or protein incorporation
US20040086544A1 (en) 1999-09-30 2004-05-06 Chienna B.V. Polymers with bioactive agents
US20040185170A1 (en) 2003-03-21 2004-09-23 Shubha Chungi Method for coating drug-containing particles and formulations and dosage units formed therefrom
WO2004084871A1 (en) 2003-03-26 2004-10-07 Ltt Bio-Pharma Co., Ltd. Intravenous nanoparticles for targenting drug delivery and sustained drug release
US20040220081A1 (en) 2002-10-30 2004-11-04 Spherics, Inc. Nanoparticulate bioactive agents
US20040219224A1 (en) 2001-06-21 2004-11-04 Kirill Yakovlevsky Spherical protein particles and methods for making and using them
US20040247680A1 (en) 2003-06-06 2004-12-09 Farokhzad Omid C. Targeted delivery of controlled release polymer systems
US20040247624A1 (en) 2003-06-05 2004-12-09 Unger Evan Charles Methods of making pharmaceutical formulations for the delivery of drugs having low aqueous solubility
US6841547B2 (en) 2003-02-28 2005-01-11 Albert Einstein College Of Medicine Of Yeshevia University Method for decreasing low density lipoprotein
US20050037086A1 (en) 1999-11-19 2005-02-17 Zycos Inc., A Delaware Corporation Continuous-flow method for preparing microparticles
US20050063976A1 (en) 1996-05-15 2005-03-24 Altarex --Medical-- Corp. Combination therapy for treating disease
US6875886B2 (en) 2001-02-07 2005-04-05 Beth Israel Deaconess Medical Center, Inc. Modified PSMA ligands and uses related thereto
US6890950B2 (en) 2002-04-23 2005-05-10 Case Western Reserve University Lapachone delivery systems, compositions and uses related thereto
US6902743B1 (en) 1995-05-22 2005-06-07 The United States Of America As Represented By The Secretary Of The Army Therapeutic treatment and prevention of infections with a bioactive material(s) encapuslated within a biodegradable-bio-compatable polymeric matrix
US20050123617A1 (en) 2001-08-31 2005-06-09 Chang Thomas M. Biodegradable polymeric nanocapsules and uses thereof
US20050136258A1 (en) 2003-12-22 2005-06-23 Shuming Nie Bioconjugated nanostructures, methods of fabrication thereof, and methods of use thereof
US20050142205A1 (en) 2003-07-18 2005-06-30 Julia Rashba-Step Methods for encapsulating small spherical particles prepared by controlled phase separation
US6916788B2 (en) 1999-12-22 2005-07-12 Samyang Corporation Liquid composition of biodegradable block copolymer for drug delivery system and process for the preparation thereof
US20050201972A1 (en) 2003-10-10 2005-09-15 Samyang Corporation Amphiphilic block copolymer and polymeric composition comprising the same for drug delivery
US20050256071A1 (en) 2003-07-15 2005-11-17 California Institute Of Technology Inhibitor nucleic acids
US20050266067A1 (en) 2004-03-02 2005-12-01 Shiladitya Sengupta Nanocell drug delivery system
US20060002971A1 (en) 2004-07-01 2006-01-05 Yale University Methods of treatment with drug loaded polymeric materials
US20060057219A1 (en) 2002-05-24 2006-03-16 Nanocarrier Co., Ltd. Method for preparing a polymer micelle pharmaceutical preparation containing drug for injection
US20060110460A1 (en) 2001-10-10 2006-05-25 Eulalia Ferret Prolonged release biodegradable microspheres and method for preparing same
US20060165987A1 (en) 2002-04-05 2006-07-27 Patrice Hildgen Stealthy polymeric biodegradable nanospheres and uses thereof
WO2006093991A1 (en) 2005-03-02 2006-09-08 The Cleveland Clinic Foundation Compounds which bind psma and uses thereof
US20070031402A1 (en) 2005-08-03 2007-02-08 Immunogen Inc. Immunoconjugate formulations
US20070043066A1 (en) 2005-07-11 2007-02-22 Wyeth Glutamate aggrecanase inhibitors
US20070041901A1 (en) 2002-06-18 2007-02-22 Diener John L Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
WO2007024323A2 (en) 2005-06-17 2007-03-01 The University Of North Carolina At Chapel Hill Nanoparticle fabrication methods, systems, and materials
US20070053845A1 (en) 2004-03-02 2007-03-08 Shiladitya Sengupta Nanocell drug delivery system
WO2007028341A1 (en) 2005-09-09 2007-03-15 Beijing Diacrid Medical Technology Co., Ltd. Nano anticancer micelles of vinca alkaloids entrapped in polyethylene glycolylated phospholipids
WO2007034479A2 (en) 2005-09-20 2007-03-29 Yissum Research Development Company Nanoparticles for targeted delivery of active agents
US20070154554A1 (en) 2005-12-29 2007-07-05 Robert Burgermeister Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
WO2007074604A1 (en) 2005-12-26 2007-07-05 Ltt Bio-Pharma Co., Ltd. Nanoparticle containing water-soluble non-peptidic low molecular weight substance
WO2007110152A2 (en) 2006-03-24 2007-10-04 Lts Lohmann Therapie-Systeme Ag Polylactide nanoparticles
CN101053553A (en) 2007-03-16 2007-10-17 吉林大学 Biodegradable fluorourcacil polyester medicine-carried nanospheres and its preparation method
WO2007133807A2 (en) 2006-05-15 2007-11-22 Massachusetts Institute Of Technology Polymers for functional particles
WO2008019142A2 (en) 2006-08-04 2008-02-14 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20080057102A1 (en) 2006-08-21 2008-03-06 Wouter Roorda Methods of manufacturing medical devices for controlled drug release
WO2008058192A2 (en) 2006-11-08 2008-05-15 Molecular Insight Pharmaceuticals, Inc. Heterodimers of glutamic acid
US20080124400A1 (en) 2004-06-24 2008-05-29 Angiotech International Ag Microparticles With High Loadings Of A Bioactive Agent
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
WO2008121949A1 (en) 2007-03-30 2008-10-09 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
WO2008124632A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
WO2008139804A1 (en) 2007-05-14 2008-11-20 Ltt Bio-Pharma Co., Ltd. Low-molecule drug-containing nanoparticle having sustained release negatively charged group
US20090053315A1 (en) 2007-08-21 2009-02-26 Board Of Regents, The University Of Texas System Thermo-Kinetic Mixing for Pharmaceutical Applications
US20090061009A1 (en) 2007-08-29 2009-03-05 Joseph Schwarz Composition and Method of Treatment of Bacterial Infections
US20090074753A1 (en) 2004-10-14 2009-03-19 Lynch Samuel E Platelet-derived growth factor compositions and methods of use thereof
EA011594B1 (en) 2004-12-30 2009-04-28 Синвеншен Аг Combination comprising an agent providing a signal, an implant material and a drug
RU2007140909A (en) 2005-04-04 2009-05-20 Синексус, Инк. (Us) DEVICE AND METHODS FOR TREATING DISEASES OF THE NANOLAIN SINUS
US20090155349A1 (en) 2006-02-23 2009-06-18 Jonathan Daniel Heller Methods of treating influenza viral infections
US20090170753A1 (en) 2006-07-26 2009-07-02 Christian Welz Capsofungin formulations
WO2009084801A1 (en) 2007-12-31 2009-07-09 Samyang Corporation Amphiphilic block copolymer micelle composition containing taxane and manufacturing process of the same
EP2106806A1 (en) 2008-03-31 2009-10-07 Fraunhofer-Gesellschaft zur Förderung der Angewandten Forschung e.V. Nanoparticles for targeted delivery of active agents to the lung
US20090306120A1 (en) 2007-10-23 2009-12-10 Florencia Lim Terpolymers containing lactide and glycolide
US20100008998A1 (en) 2006-09-26 2010-01-14 Samyang Corporation Submicron nanoparticle of poorly water soluble camptothecin derivatives and process for preparation thereof
US20100015050A1 (en) 2006-12-21 2010-01-21 Wayne State University Peg and targeting ligands on nanoparticle surface
US20100040537A1 (en) 2008-07-08 2010-02-18 Abbott Laboratories Prostaglandin E2 Binding Proteins and Uses Thereof
US20100069426A1 (en) 2008-06-16 2010-03-18 Zale Stephen E Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US20100068286A1 (en) 2008-06-16 2010-03-18 Greg Troiano Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20100087337A1 (en) 2008-09-10 2010-04-08 Bind Biosciences, Inc. High Throughput Fabrication of Nanoparticles
US20100104655A1 (en) 2008-06-16 2010-04-29 Zale Stephen E Therapeutic Polymeric Nanoparticles Comprising Vinca Alkaloids and Methods of Making and Using Same
US20100104645A1 (en) 2008-06-16 2010-04-29 Bind Biosciences, Inc. Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic targeted nanoparticles
WO2010068866A2 (en) 2008-12-12 2010-06-17 Bind Biosciences Therapeutic particles suitable for parenteral administration and methods of making and using same
US20100166866A1 (en) 2003-03-26 2010-07-01 Egalet A/S Matrix compositions for controlled delivery of drug substances
US20100216804A1 (en) 2008-12-15 2010-08-26 Zale Stephen E Long Circulating Nanoparticles for Sustained Release of Therapeutic Agents
WO2010114770A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-agent conjugates, particles, compositions, and related methods of use
WO2010114768A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-epothilone conjugates, particles, compositions, and related methods of use
US20100303900A1 (en) 2000-05-25 2010-12-02 Alkermes Controlled Therapeutics, Inc. Preparation of injectable suspensions having improved injectability
US20100303723A1 (en) 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
US20100316725A1 (en) 2009-05-27 2010-12-16 Elan Pharma International Ltd. Reduction of flake-like aggregation in nanoparticulate active agent compositions
WO2011079279A2 (en) 2009-12-23 2011-06-30 Board Of Trustees Of The University Of Illinois Nanoconjugates and nanoconjugate formulations
US20110159079A1 (en) 2002-10-29 2011-06-30 Zhili Li High Delivery Rates for Lipid Based Drug Formulations, and Methods of Treatment Thereof
WO2011084521A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
WO2011084518A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
WO2011084513A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
US8034765B2 (en) 2005-08-31 2011-10-11 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US20110275704A1 (en) 2009-12-11 2011-11-10 Greg Troiano Stable Formulations for Lyophilizing Therapeutic Particles
WO2012054923A2 (en) 2010-10-22 2012-04-26 Bind Biosciences, Inc. Therapeutic nanoparticles with high molecular weight copolymers
WO2012166923A2 (en) 2011-05-31 2012-12-06 Bind Biosciences Drug loaded polymeric nanoparticles and methods of making and using same
WO2013044219A1 (en) 2011-09-22 2013-03-28 Bind Biosciences Methods of treating cancers with therapeutic nanoparticles

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0394265B1 (en) 1987-07-29 1994-11-02 The Liposome Company, Inc. Method for size separation of particles
US5542935A (en) 1989-12-22 1996-08-06 Imarx Pharmaceutical Corp. Therapeutic delivery systems related applications
GB9412273D0 (en) 1994-06-18 1994-08-10 Univ Nottingham Administration means
US5792477A (en) 1996-05-07 1998-08-11 Alkermes Controlled Therapeutics, Inc. Ii Preparation of extended shelf-life biodegradable, biocompatible microparticles containing a biologically active agent
JP2942508B2 (en) 1997-01-14 1999-08-30 順也 藤森 Temperature sensitive sustained release base and temperature sensitive sustained release system
CA2335852A1 (en) 1998-06-30 2000-01-06 Subodh Shah Thermosensitive biodegradable hydrogels for sustained delivery of biologically active agents
KR100274842B1 (en) 1998-10-01 2001-03-02 김효근 Sustained-release Drug Release System of Retinoic Acid Using Microspheres
DE19856432A1 (en) 1998-12-08 2000-06-15 Basf Ag Nanoparticulate core-shell systems and their use in pharmaceutical and cosmetic preparations
US6194006B1 (en) 1998-12-30 2001-02-27 Alkermes Controlled Therapeutics Inc. Ii Preparation of microparticles having a selected release profile
US6890946B2 (en) 1999-12-23 2005-05-10 Indiana University Research And Technology Corporation Use of parthenolide to inhibit cancer
KR100446101B1 (en) 2000-12-07 2004-08-30 주식회사 삼양사 Sustained delivery composition for poorly water soluble drugs
JP2004516262A (en) 2000-12-21 2004-06-03 ネクター セラピューティクス Induced phase transition method for the production of microparticles containing hydrophilic activators
AU2002338336A1 (en) 2001-04-03 2002-10-21 Kosan Biosciences, Inc. Epothilone derivatives and methods for making and using the same
EP1478343B1 (en) 2001-10-15 2017-03-22 Crititech, Inc. Compositions and methods for delivery of poorly water soluble drugs and methods of treatment
US20040126400A1 (en) * 2002-05-03 2004-07-01 Iversen Patrick L. Delivery of therapeutic compounds via microparticles or microbubbles
EP1578193A4 (en) 2002-12-23 2011-06-15 Vical Inc Method for freeze-drying nucleic acid/block copolymer/cationic surfactant complexes
AU2004228008B2 (en) 2003-04-03 2008-11-06 Jessie L.-S. Au Tumor-targeting drug-loaded particles
ES2741576T3 (en) 2003-07-23 2020-02-11 Evonik Corp Controlled release compositions
JP2007504259A (en) 2003-09-02 2007-03-01 ノバルティス アクチエンゲゼルシャフト Cancer treatment with epothilone
US8043631B2 (en) 2004-04-02 2011-10-25 Au Jessie L S Tumor targeting drug-loaded particles
JP2006131577A (en) 2004-11-09 2006-05-25 Ltt Bio-Pharma Co Ltd Method for preparing nanoparticle having different particle diameters and containing sealed medicine and nanoparticle obtained by the method
JP2006321763A (en) 2005-05-20 2006-11-30 Hosokawa Funtai Gijutsu Kenkyusho:Kk Biocompatibilie nanoparticle and method for production of the same
US20090022806A1 (en) 2006-12-22 2009-01-22 Mousa Shaker A Nanoparticle and polymer formulations for thyroid hormone analogs, antagonists and formulations and uses thereof
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
EP2056793A4 (en) 2006-07-31 2011-08-17 Neurosystec Corp Free base gacyclidine nanoparticles
CN1957911A (en) 2006-12-01 2007-05-09 济南康泉医药科技有限公司 Controlled release formulation for anti entity tumour
CN101396340A (en) 2006-12-12 2009-04-01 济南帅华医药科技有限公司 Anti-cancer sustained-released injection containing epothilone derivate
CN101396342A (en) 2006-12-12 2009-04-01 济南帅华医药科技有限公司 Anti-cancer sustained-released injection containing epothilone derivate
CN1969818A (en) 2006-12-12 2007-05-30 济南帅华医药科技有限公司 Anticancer sustained release injection containing epothilone derivatives
CN101433520A (en) 2006-12-12 2009-05-20 济南帅华医药科技有限公司 Anticancer sustained-release agent containing epothilone
CN1969816A (en) 2006-12-12 2007-05-30 济南帅华医药科技有限公司 Anticancer sustained release agent containing epothilone
CN1961864A (en) 2006-12-12 2007-05-16 济南帅华医药科技有限公司 Anticancer composition
CA3201293A1 (en) 2007-03-07 2008-09-12 Abraxis Bioscience, Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
US9422234B2 (en) 2007-11-30 2016-08-23 The Johns Hopkins University Prostate specific membrane antigen (PSMA) targeted nanoparticles for therapy of prostate cancer
DE102007059752A1 (en) 2007-12-10 2009-06-18 Bayer Schering Pharma Aktiengesellschaft Functionalized solid polymer nanoparticles containing epothilones
EA201171195A8 (en) 2009-03-30 2014-08-29 Серулин Фарма Инк. CONJUGATES, PARTICLES, POLYMER-AGENT COMPOSITIONS AND METHODS OF THEIR APPLICATION
WO2012040513A1 (en) 2010-09-22 2012-03-29 The Board Of Regents Of The University Of Texas System Compositions and methods for the delivery of beta lapachone
CA2865700C (en) 2012-02-29 2020-05-05 Merck Patent Gmbh Process for the production of nanoparticles laden with active compound
AU2013315125B2 (en) 2012-09-17 2018-07-26 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using same
JP6356678B2 (en) 2012-09-17 2018-07-11 ファイザー・インク Method for producing therapeutic nanoparticles
MX2016000131A (en) 2013-06-28 2016-06-15 Bind Therapeutics Inc Docetaxel polymeric nanoparticles for cancer treatment.

Patent Citations (142)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766635A (en) 1991-06-28 1998-06-16 Rhone-Poulenc Rorer S.A. Process for preparing nanoparticles
US5563122A (en) 1991-12-09 1996-10-08 Asahi Kasei Kogyo Kabushiki Kaisha Stabilized parathyroid hormone composition
US5302401A (en) 1992-12-09 1994-04-12 Sterling Winthrop Inc. Method to reduce particle size growth during lyophilization
US5543158A (en) * 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5578325A (en) 1993-07-23 1996-11-26 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
EP0805678A1 (en) 1995-01-05 1997-11-12 THE BOARD OF REGENTS acting for and on behalf of THE UNIVERSITY OF MICHIGAN Surface-modified nanoparticles and method of making and using same
US6346274B1 (en) 1995-03-10 2002-02-12 Roche Diagnostics Gmbh Polypeptide-containing pharmaceutical forms of administration in the form of microparticles and method for the preparation thereof
US6902743B1 (en) 1995-05-22 2005-06-07 The United States Of America As Represented By The Secretary Of The Army Therapeutic treatment and prevention of infections with a bioactive material(s) encapuslated within a biodegradable-bio-compatable polymeric matrix
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US6139870A (en) 1995-12-19 2000-10-31 Aventis Pharma Sa Stabilized nanoparticles which are filterable under sterile conditions
US20050063976A1 (en) 1996-05-15 2005-03-24 Altarex --Medical-- Corp. Combination therapy for treating disease
US6201072B1 (en) 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6254890B1 (en) 1997-12-12 2001-07-03 Massachusetts Institute Of Technology Sub-100nm biodegradable polymer spheres capable of transporting and releasing nucleic acids
US20020045582A1 (en) 1997-12-31 2002-04-18 Alexey L. Margolin Stabilized protein crystals formulations containing them and methods of making them
US6265609B1 (en) 1998-07-06 2001-07-24 Guilford Pharmaceuticals Inc. Thio-substituted pentanedioic acid derivatives
US6395718B1 (en) 1998-07-06 2002-05-28 Guilford Pharmaceuticals Inc. Pharmaceutical compositions and methods of inhibiting angiogenesis using naaladase inhibitors
US20040071768A1 (en) 1999-04-01 2004-04-15 Inex Pharmaceuticals Corporation Compositions and methods for treating cancer
US20040086544A1 (en) 1999-09-30 2004-05-06 Chienna B.V. Polymers with bioactive agents
US6136846A (en) 1999-10-25 2000-10-24 Supergen, Inc. Formulation for paclitaxel
US20050037086A1 (en) 1999-11-19 2005-02-17 Zycos Inc., A Delaware Corporation Continuous-flow method for preparing microparticles
US6916788B2 (en) 1999-12-22 2005-07-12 Samyang Corporation Liquid composition of biodegradable block copolymer for drug delivery system and process for the preparation thereof
US6528499B1 (en) 2000-04-27 2003-03-04 Georgetown University Ligands for metabotropic glutamate receptors and inhibitors of NAALADase
US20030143184A1 (en) 2000-05-17 2003-07-31 Min-Hyo Seo Stable polymeric micelle-type drug composition and method for the preparation thereof
US20100303900A1 (en) 2000-05-25 2010-12-02 Alkermes Controlled Therapeutics, Inc. Preparation of injectable suspensions having improved injectability
KR20020041712A (en) 2000-11-28 2002-06-03 윤덕용 Process for Preparing Sustained Release Form of Micelle Employing Conjugate of Anticancer Drug and Biodegradable Polymer
KR100418916B1 (en) 2000-11-28 2004-02-14 한국과학기술원 Process for Preparing Sustained Release Form of Micelle Employing Conjugate of Anticancer Drug and Biodegradable Polymer
US20020119916A1 (en) 2000-12-22 2002-08-29 Hassan Emadeldin M. Elemental nanoparticles of substantially water insoluble materials
US20040081688A1 (en) 2000-12-27 2004-04-29 Del Curto Maria Dorly Amphiphilic lipid nanoparticles for peptide and/or protein incorporation
US6875886B2 (en) 2001-02-07 2005-04-05 Beth Israel Deaconess Medical Center, Inc. Modified PSMA ligands and uses related thereto
US20040219224A1 (en) 2001-06-21 2004-11-04 Kirill Yakovlevsky Spherical protein particles and methods for making and using them
US20050123617A1 (en) 2001-08-31 2005-06-09 Chang Thomas M. Biodegradable polymeric nanocapsules and uses thereof
US20030068377A1 (en) 2001-10-03 2003-04-10 Macromed, Incorporated PLA/PLGA oligomers combined with block copolymers for enhancing solubility of a drug in water
US20060110460A1 (en) 2001-10-10 2006-05-25 Eulalia Ferret Prolonged release biodegradable microspheres and method for preparing same
US20030235619A1 (en) 2001-12-21 2003-12-25 Christine Allen Polymer-lipid delivery vehicles
WO2003055469A1 (en) 2001-12-21 2003-07-10 Celator Technologies Inc. Improved polymer-lipid delivery vehicles
US20040054190A1 (en) 2002-01-10 2004-03-18 Johns Hopkins University Imaging agents and methods of imaging NAALADase or PSMA
US20060165987A1 (en) 2002-04-05 2006-07-27 Patrice Hildgen Stealthy polymeric biodegradable nanospheres and uses thereof
US20030232887A1 (en) 2002-04-10 2003-12-18 Johnson Douglas Giles Preparation and use of a stable formulation of allosteric effector compounds
US6890950B2 (en) 2002-04-23 2005-05-10 Case Western Reserve University Lapachone delivery systems, compositions and uses related thereto
US20060057219A1 (en) 2002-05-24 2006-03-16 Nanocarrier Co., Ltd. Method for preparing a polymer micelle pharmaceutical preparation containing drug for injection
US20070041901A1 (en) 2002-06-18 2007-02-22 Diener John L Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
US20110159079A1 (en) 2002-10-29 2011-06-30 Zhili Li High Delivery Rates for Lipid Based Drug Formulations, and Methods of Treatment Thereof
US20040220081A1 (en) 2002-10-30 2004-11-04 Spherics, Inc. Nanoparticulate bioactive agents
US6841547B2 (en) 2003-02-28 2005-01-11 Albert Einstein College Of Medicine Of Yeshevia University Method for decreasing low density lipoprotein
US20040185170A1 (en) 2003-03-21 2004-09-23 Shubha Chungi Method for coating drug-containing particles and formulations and dosage units formed therefrom
US20100166866A1 (en) 2003-03-26 2010-07-01 Egalet A/S Matrix compositions for controlled delivery of drug substances
WO2004084871A1 (en) 2003-03-26 2004-10-07 Ltt Bio-Pharma Co., Ltd. Intravenous nanoparticles for targenting drug delivery and sustained drug release
US20040247624A1 (en) 2003-06-05 2004-12-09 Unger Evan Charles Methods of making pharmaceutical formulations for the delivery of drugs having low aqueous solubility
US20050037075A1 (en) * 2003-06-06 2005-02-17 Farokhzad Omid C. Targeted delivery of controlled release polymer systems
US20040247680A1 (en) 2003-06-06 2004-12-09 Farokhzad Omid C. Targeted delivery of controlled release polymer systems
WO2005046572A2 (en) 2003-06-06 2005-05-26 Massachusetts Institute Of Technology Targeted delivery of controlled release polymer systems
US20050256071A1 (en) 2003-07-15 2005-11-17 California Institute Of Technology Inhibitor nucleic acids
US20050142205A1 (en) 2003-07-18 2005-06-30 Julia Rashba-Step Methods for encapsulating small spherical particles prepared by controlled phase separation
US20050201972A1 (en) 2003-10-10 2005-09-15 Samyang Corporation Amphiphilic block copolymer and polymeric composition comprising the same for drug delivery
US20050136258A1 (en) 2003-12-22 2005-06-23 Shuming Nie Bioconjugated nanostructures, methods of fabrication thereof, and methods of use thereof
US20070053845A1 (en) 2004-03-02 2007-03-08 Shiladitya Sengupta Nanocell drug delivery system
US20050266067A1 (en) 2004-03-02 2005-12-01 Shiladitya Sengupta Nanocell drug delivery system
US20080124400A1 (en) 2004-06-24 2008-05-29 Angiotech International Ag Microparticles With High Loadings Of A Bioactive Agent
US20060002971A1 (en) 2004-07-01 2006-01-05 Yale University Methods of treatment with drug loaded polymeric materials
US20060002852A1 (en) 2004-07-01 2006-01-05 Yale University Targeted and high density drug loaded polymeric materials
US20090074753A1 (en) 2004-10-14 2009-03-19 Lynch Samuel E Platelet-derived growth factor compositions and methods of use thereof
EA011594B1 (en) 2004-12-30 2009-04-28 Синвеншен Аг Combination comprising an agent providing a signal, an implant material and a drug
WO2006093991A1 (en) 2005-03-02 2006-09-08 The Cleveland Clinic Foundation Compounds which bind psma and uses thereof
RU2007140909A (en) 2005-04-04 2009-05-20 Синексус, Инк. (Us) DEVICE AND METHODS FOR TREATING DISEASES OF THE NANOLAIN SINUS
WO2007024323A2 (en) 2005-06-17 2007-03-01 The University Of North Carolina At Chapel Hill Nanoparticle fabrication methods, systems, and materials
US20070043066A1 (en) 2005-07-11 2007-02-22 Wyeth Glutamate aggrecanase inhibitors
US20070031402A1 (en) 2005-08-03 2007-02-08 Immunogen Inc. Immunoconjugate formulations
US8034765B2 (en) 2005-08-31 2011-10-11 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
WO2007028341A1 (en) 2005-09-09 2007-03-15 Beijing Diacrid Medical Technology Co., Ltd. Nano anticancer micelles of vinca alkaloids entrapped in polyethylene glycolylated phospholipids
US20090053293A1 (en) 2005-09-09 2009-02-26 Beijing Diacrid Medical Technology Co., Ltd. Nano Anticancer Micelles of Vinca Alkaloids Entrapped in Polyethylene Glycolylated Phospholipids
WO2007034479A2 (en) 2005-09-20 2007-03-29 Yissum Research Development Company Nanoparticles for targeted delivery of active agents
US20080267876A1 (en) 2005-09-20 2008-10-30 Yissum Research Development Company Nanoparticles for Targeted Delivery of Active Agent
WO2007074604A1 (en) 2005-12-26 2007-07-05 Ltt Bio-Pharma Co., Ltd. Nanoparticle containing water-soluble non-peptidic low molecular weight substance
US20090317479A1 (en) 2005-12-26 2009-12-24 Tsutomu Ishihara Nanoparticles containing water-soluble non-peptide low-molecular weight drug
EP1985309A1 (en) 2005-12-26 2008-10-29 LTT Bio-Pharma Co., Ltd. Nanoparticle containing water-soluble non-peptidic low molecular weight substance
US20070154554A1 (en) 2005-12-29 2007-07-05 Robert Burgermeister Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
US20090155349A1 (en) 2006-02-23 2009-06-18 Jonathan Daniel Heller Methods of treating influenza viral infections
US8003128B2 (en) 2006-03-24 2011-08-23 Lts Lohmann Therapie-Systeme Ag Polylactide nanoparticles
WO2007110152A2 (en) 2006-03-24 2007-10-04 Lts Lohmann Therapie-Systeme Ag Polylactide nanoparticles
US20100266491A1 (en) 2006-03-31 2010-10-21 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US20080081074A1 (en) 2006-05-15 2008-04-03 Massachusetts Institute Of Technology Polymers for functional particles
WO2007133807A2 (en) 2006-05-15 2007-11-22 Massachusetts Institute Of Technology Polymers for functional particles
US20090170753A1 (en) 2006-07-26 2009-07-02 Christian Welz Capsofungin formulations
WO2008019142A2 (en) 2006-08-04 2008-02-14 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20080057102A1 (en) 2006-08-21 2008-03-06 Wouter Roorda Methods of manufacturing medical devices for controlled drug release
US20100008998A1 (en) 2006-09-26 2010-01-14 Samyang Corporation Submicron nanoparticle of poorly water soluble camptothecin derivatives and process for preparation thereof
WO2008058192A2 (en) 2006-11-08 2008-05-15 Molecular Insight Pharmaceuticals, Inc. Heterodimers of glutamic acid
US20080193381A1 (en) 2006-11-08 2008-08-14 Molecular Insight Pharmaceuticals, Inc. Heterodimers of glutamic acid
US20100303723A1 (en) 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
US20100015050A1 (en) 2006-12-21 2010-01-21 Wayne State University Peg and targeting ligands on nanoparticle surface
CN101053553A (en) 2007-03-16 2007-10-17 吉林大学 Biodegradable fluorourcacil polyester medicine-carried nanospheres and its preparation method
WO2008121949A1 (en) 2007-03-30 2008-10-09 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
US20090061010A1 (en) 2007-03-30 2009-03-05 Massachusetts Institute Of Technology Cancer cell targeting using nanoparticles
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
WO2008124639A2 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
WO2008124632A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
US20090074828A1 (en) 2007-04-04 2009-03-19 Massachusetts Institute Of Technology Poly(amino acid) targeting moieties
WO2008139804A1 (en) 2007-05-14 2008-11-20 Ltt Bio-Pharma Co., Ltd. Low-molecule drug-containing nanoparticle having sustained release negatively charged group
US20090053315A1 (en) 2007-08-21 2009-02-26 Board Of Regents, The University Of Texas System Thermo-Kinetic Mixing for Pharmaceutical Applications
US20090061009A1 (en) 2007-08-29 2009-03-05 Joseph Schwarz Composition and Method of Treatment of Bacterial Infections
US20110224288A1 (en) 2007-09-28 2011-09-15 Zale Stephen E Cancer Cell Targeting Using Nanoparticles
US20120004293A1 (en) 2007-09-28 2012-01-05 Zale Stephen E Cancer Cell Targeting Using Nanoparticles
US20090306120A1 (en) 2007-10-23 2009-12-10 Florencia Lim Terpolymers containing lactide and glycolide
WO2009084801A1 (en) 2007-12-31 2009-07-09 Samyang Corporation Amphiphilic block copolymer micelle composition containing taxane and manufacturing process of the same
EP2106806A1 (en) 2008-03-31 2009-10-07 Fraunhofer-Gesellschaft zur Förderung der Angewandten Forschung e.V. Nanoparticles for targeted delivery of active agents to the lung
US8318211B2 (en) 2008-06-16 2012-11-27 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US8318208B1 (en) 2008-06-16 2012-11-27 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US20120276162A1 (en) 2008-06-16 2012-11-01 Zale Stephen E Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20110274759A1 (en) 2008-06-16 2011-11-10 Greg Troiano Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20100069426A1 (en) 2008-06-16 2010-03-18 Zale Stephen E Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
US20100068286A1 (en) 2008-06-16 2010-03-18 Greg Troiano Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20100104645A1 (en) 2008-06-16 2010-04-29 Bind Biosciences, Inc. Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic targeted nanoparticles
US20100068285A1 (en) 2008-06-16 2010-03-18 Zale Stephen E Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US8206747B2 (en) 2008-06-16 2012-06-26 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US20100104655A1 (en) 2008-06-16 2010-04-29 Zale Stephen E Therapeutic Polymeric Nanoparticles Comprising Vinca Alkaloids and Methods of Making and Using Same
US20100040537A1 (en) 2008-07-08 2010-02-18 Abbott Laboratories Prostaglandin E2 Binding Proteins and Uses Thereof
US20100087337A1 (en) 2008-09-10 2010-04-08 Bind Biosciences, Inc. High Throughput Fabrication of Nanoparticles
WO2010068866A2 (en) 2008-12-12 2010-06-17 Bind Biosciences Therapeutic particles suitable for parenteral administration and methods of making and using same
US20100226986A1 (en) 2008-12-12 2010-09-09 Amy Grayson Therapeutic Particles Suitable for Parenteral Administration and Methods of Making and Using Same
US20130034608A1 (en) 2008-12-15 2013-02-07 Zale Stephen E Long Circulating Nanoparticles for Sustained Release of Therapeutic Agents
US20110217377A1 (en) 2008-12-15 2011-09-08 Zale Stephen E Long Circulating Nanoparticles for Sustained Release of Therapeutic Agents
US20100216804A1 (en) 2008-12-15 2010-08-26 Zale Stephen E Long Circulating Nanoparticles for Sustained Release of Therapeutic Agents
WO2010114770A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-agent conjugates, particles, compositions, and related methods of use
WO2010114768A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-epothilone conjugates, particles, compositions, and related methods of use
US20100316725A1 (en) 2009-05-27 2010-12-16 Elan Pharma International Ltd. Reduction of flake-like aggregation in nanoparticulate active agent compositions
US20110275704A1 (en) 2009-12-11 2011-11-10 Greg Troiano Stable Formulations for Lyophilizing Therapeutic Particles
US8357401B2 (en) 2009-12-11 2013-01-22 Bind Biosciences, Inc. Stable formulations for lyophilizing therapeutic particles
US8211473B2 (en) 2009-12-11 2012-07-03 Bind Biosciences, Inc. Stable formulations for lyophilizing therapeutic particles
WO2011084521A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
US20120140790A1 (en) 2009-12-15 2012-06-07 Ali Mir M Therapeutic Polymeric Nanoparticle Compositions with High Glass Transition Termperature or High Molecular Weight Copolymers
US20110294717A1 (en) 2009-12-15 2011-12-01 Ali Mir M Therapeutic Polymeric Nanoparticle Compositions with High Glass Transition Temperature or High Molecular Weight Copolymers
WO2011084513A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
WO2011084518A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
US20130108668A1 (en) 2009-12-15 2013-05-02 Maria Figueiredo Therapeutic Polymeric Nanoparticles Comprising Epothilone and Methods of Making and Using Same
US20130115293A1 (en) 2009-12-15 2013-05-09 Abhimanyu Sabnis Therapeutic Polymeric Nanoparticles Comprising Corticosteroids and Methods of Making and Using Same
WO2011079279A2 (en) 2009-12-23 2011-06-30 Board Of Trustees Of The University Of Illinois Nanoconjugates and nanoconjugate formulations
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
WO2012054923A2 (en) 2010-10-22 2012-04-26 Bind Biosciences, Inc. Therapeutic nanoparticles with high molecular weight copolymers
WO2012166923A2 (en) 2011-05-31 2012-12-06 Bind Biosciences Drug loaded polymeric nanoparticles and methods of making and using same
WO2013044219A1 (en) 2011-09-22 2013-03-28 Bind Biosciences Methods of treating cancers with therapeutic nanoparticles

Non-Patent Citations (99)

* Cited by examiner, † Cited by third party
Title
"Docetaxel Dosage," [retrieved on Mar. 28, 2013] http://www.drugs.com/dosage/docetaxel.html.
"Taxotere Dosage," [retrieved on Mar. 28, 2013]. http://www.drugs.com/dosage/taxotere.html.c.
Abdelwahed et al., "Freeze-Drying of Nanoparticles: Formulation, Process and Storage Considerations," Adv. Drug Deliv. Rev. (2006) 58:1688-1713.
Abizaid, A. et al., "Sirolimus-Eluting Stents Inhibit Neointimal Hyperplasia in Diabetic Patients," Euro. Heart J. (2006) 25:104-112.
Adams, M. et al., "Amphiphilic Block Copolymers for Drug Delivery," J. Pharm. Sci. (2003) 92(7):1343-1355.
Altmann (Epothilone B and its analogs-a new family of anticancer agents, Mini Rev Med. Chem. (2003) 3(2):149-158; Abstract Only [retrieved from Bentham Science, ], 1 page.
Altmann (Epothilone B and its analogs—a new family of anticancer agents, Mini Rev Med. Chem. (2003) 3(2):149-158; Abstract Only [retrieved from Bentham Science, < URL: http://www.eurekaselect.com/80911/artcile>], 1 page.
Altmann (Epothilone B and its analogs-a new family of anticancer agents, Mini Rev Med.Chem. (2003) 3(2):149-158; Abstract Only [retrieved from PUBMED, ]), 1 page.
Altmann (Epothilone B and its analogs—a new family of anticancer agents, Mini Rev Med.Chem. (2003) 3(2):149-158; Abstract Only [retrieved from PUBMED, < URL: http://www.ncbi.nlm.nih.gov/pubmed/12570848>]), 1 page.
Avgoustakis, "Pegylated poly(lactide) and poly(lactide-co-glycolide) nanoparticles: Preparation, properties and possible applications in drug delivery," Current Drug Delivery. (2004) 1(4):321-333.
Barinka, Cyril, et al., "Interactions Between Human Glutamate Carboxypeptidase II and Urea-Based Inhibitors: Structural Characterization," J. Med. Chem. 2008, 51, 7737-7743, pp. 7737-7743.
Barinka, Cyril, et al., Structural Insight into the Pharmacophore Pocket of Human Glutamate Carboxypeptidase II, J. Med. Chem. 2007, 50, pp. 3267-3273.
Bilati et al., "Nanoprecipitation Versus Emulsion-based Techniques for the Encapsulation of Proteins into Biodegradable Nanoparticles and Process-related Stability Issues " AAPS PharmSciTech. (2005) 6(4):E594-E604.
Blindt, Rudiger et al., "A Novel Drug-Eluting Stent Coated with an Integrin-Binding Cyclic Arg-Gly-Asp Peptide Inhibits Neointimal Hyperplasia by Recruiting Endothelial Progenitor Cells," J. Am. Coll. Cardiol. (2006) 47(9):1786-1795.
Caliceti, P. et al., "Effective Protein Release from PEG/PLA Nano-Particles Produced by Compressed Gas Anti-Solvent Precipitation Techniques," J. Control. Rel. (2004) 94:195-205.
Chandran, Sachin S., et al., "Characterization of a Targeted Nanoparticle Functionalized with a Urea-Based Inhibitor of Prostate-Specific Membrane Antigen (PSMA)," Cancer Biology & Therapy 7:4, pp. 1-9; Apr. 2008.
Chen, Ying, et al., "Radiohalogenated Prostate-Specific Membrane Antigen (PSMA)-Based Ureas as Imaging Agents for Prostate Cancer," J. Med. Chem., 2008, 51 (24), pp. 7933-7943.
CR Heald, S Stolnick, KS Kujawinski, C De Matteis, MC Garnett, L Illum, SS Davis, SC Purkiss, RJ Barlow, PR Gellert. "Poly(lactic acid)-Poly(ethylene oxide) (PLA-PEG) Nanoparticles: NMR Studies of the Central Solidlike PLA Core and the Liquid PEG Corona." Langmuir, vol. 18, 2002, pp. 3669-3675. *
Davaran et al., "Preparation and in Vitro Evaluation of Linear and Star-Branched PLGA Nanoparticles for Insulin Delivery," J. Bioact. Compat. Polym. (2008) 23:115-131.
De Jaeghere et al., "Formulation and Lyoprotection of Poly(lactic acid-co-ethylene oxide) Nanoparticles: Influence on Physical Stability and in Vitro Cell Uptake " Pharm. Res. (1999) 16(6):859-866.
Eurasian Search Report for Application No. EA 201170038, dated Jul. 8, 2011.
Eurasian Search Report for Application No. EA 201170039, dated Nov. 21, 2011.
Ewesuedo et al., "Chapter 1: Systemically Administrated Drugs." Drug Delivery Systems in Cancer Therapy. Ed. D.M. Brown. Totowa:Humana, 2003, pp. 3-14.
Extended European Search Report for Application No. EP 09794915.0, mailed Jan. 25, 2012.
Extended European Search Report for Application No. EP 09835578.7, mailed May 18, 2012.
Extended European Search Report for Application No. EP 10836748.3, mailed Mar. 21, 2013.
Extended European Search Report for European Application No. 09794913.5 mailed Jul. 8, 2011.
F De Jaeghere, E Allemann, J Feijen, T Kissel, E Doelker, R Gurny. "Freeze-Drying and Lyopreservation of Diblock and Triblock Poly(Lactic Acid)-Poly(Ethylene Oxide) (PLA-PEO) Copolymer Nanoparticles." Pharmaceutical Development and Technology, vol. 5(4), 2000, pp. 473-483. *
Farokhzad, Omid C., et al., "Nanoparticle-Aptamer Bioconjugates: A New Approach for Targeting Prostate Cancer Cells," Cancer Research 64, pp. 7668-7672, Nov. 1, 2004.
Farokhzad, Omid C., et al., "Targeted Nanoparticle-Aptamer Bioconjugates for Cancer Chemotherapy in vivo," PNAS (2006) 103:16, pp. 6315-6320.
Feng et al., "Nanoparticles of Biodegradable Polymers for Clinical Administration of Paclitaxel," Current Medicinal Chemistry, 11:413-424. (2004).
Foss, Catherine A., et al., "Radiolabeled Small-Molecule Ligands for Prostate-Specific Membrane Antigen: In Vivo Imaging in Experimental Models of Prostate Cancer," Clin. Cancer Res. 2005;11(11), pp. 4022-4028.
Foss, Catherine, Poster Session: Novel Probes and Activation Strategies, Part 3, "Synthesis and Validation of a Novel Small-Molecule Fluorescent Probe for PSMA Expression in Human Tumor Neovasculature," 4th Annual Meeting for the Society for Molecular Imaging, Sep. 7-10, 2005.
Fournier et al., "Experimental Studies and Preliminary Clinical Trial of Vinorelbine-loaded Polymeric Bioresorbable Implants for the Local Treatment of Solid Tumors," Cancer Research.(1991) 51: 5384-5391.
Galsky et al., "Cabazitaxel " Nature Reviews. (2010) 9:677-678.
Gao, X. et al., "In Vivo Cancer Targeting and Imaging with Semiconductor Quantum Dots," Nat. Biotechnol. (2004) 22(8):969-976.
Govender et al., "Defining the Drug Incorporation Properties of PLA-PEG Nanoparticles," Int. J. Pharm. (2000) 199: 95-110.
Gref et al., Development and Characterization of CyA-loaded Poly(lactic acid)-poly(ethylene glycol)PEG Micro- and Nanoparticles. Comparison with Conventional PLA Particulate Carriers. Eur. J. Pharm. Biopharm. (2001) 51:111-118.
Gref, Ruxandra, et al., "Biodegradable Long-Circulating Polymeric Nanospheres," Science, 1994, vol. 263, pp. 1600-1603.
Gu, F. et al. "Precise Engineering of Targeted Nanoparticles by Using Self-Assembled Biointegrated Block Copolymers," Proc. Natl. Acad. Sci. USA. (2008) 105(7):2586-2591.
Hederstrom et al., "Purification and Surface Modification of Polymeric Nanoparticles for Medical Applications." Masters thesis. SINTEF Materials and Chemistry, Trondheim, Norway, Mar. 3, 2008.
Heldman, A. et al., "Paclitaxel Stent Coating Inhibits Neointimal Hyperplasia at 4 Weeks in a Porcine Model of Coronary Restenosis," Circulation (2001) 103:2289-2295.
Hrkach et al., "Preclinical Development and Clinical Translation of a PSMA-Targeted Docetaxel Nanoparticle with a Differentiated Pharmacological Profile " Sci. Trans. Med. (2012) 4:1-11.
Humblet, Valerie, et al., "An HPLC/Mass Spectrometry Platform for the Development of Multimodality Contrast Agents and Targeted Therapeutics: Prostate-Specific Membrane Antigen Small Molecule Derivatives," Contrast Med. Mol. Imaging 1: pp. 196-211 (2006).
Humblet, Valerie, et al., "High-Affinity Near-Infrared Fluorescent Small-Molecule Contrast Agents for In Vivo Imaging of Prostate-Specific Membrane Antigen," Molecular Imaging, vol. 4, Oct. 2005, pp. 448-462.
International Preliminary Report on Patentability for PCT/US2010/060575 dated Jun. 19, 2012, 11 pages.
International Search Report for Application No. PCT/US08/13158 dated Jan. 20, 2009 and mailed Feb. 17, 2009.
International Search Report for Application No. PCT/US08/58873 dated Aug. 15, 2008 and mailed Aug. 28, 2008.
International Search Report for Application No. PCT/US09/47513 dated Jan. 18, 2010 and mailed Jan. 18, 2010.
International Search Report for Application No. PCT/US09/67672 dated Aug. 20, 2010 and mailed Aug. 23, 2010.
International Search Report for Application No. PCT/US09/68028 dated Aug. 9, 2010 and mailed Aug. 23, 2010.
International Search Report for Application No. PCT/US10/59879 dated Aug. 30, 2011 and mailed Aug. 30, 2011.
International Search Report for Application No. PCT/US10/60564 dated Sep. 29, 2011 and mailed Sep. 29, 2011.
International Search Report for Application No. PCT/US10/60570 dated Aug. 25, 2011 and mailed Aug. 25, 2011.
International Search Report for Application No. PCT/US10/60575 dated Aug. 25, 2011 and mailed Aug. 25, 2011.
International Search Report for Application No. PCT/US11/057498 dated May 9, 2012 and mailed May 10, 2012.
International Search Report for Application No. PCT/US2012/040215 dated Nov. 16, 2012 and mailed Nov. 16, 2012.
International Search Report for Application No. PCT/US2012/056891 dated Jan. 4, 2013 and mailed Jan. 4, 2013.
International Search Report for PCT/US09/47515 dated Jan. 18, 2010 and mailed Jan. 19, 2010.
International Search Report for PCT/US09/47517 dated Feb. 23, 2010 and mailed Mar. 2, 2010.
International Search Report for PCT/US09/47518 dated Mar. 5, 2010 and mailed Mar. 5, 2010.
J Cheng, BA Teply, I Sherifi, J Sung, G Luther, FX Gu, EL Nissenbaum, AFR Moreno, R Langer, OC Farokhzad. "Formulation of Functionalized PLGA-PEG Nanoparticles for In Vivo Targeted Drug Delivery." Biomaterials, vol. 28, 2007, pp. 869-876, available online Oct. 20, 2006. *
Japaprakash, Sarva, et al., "Design and Synthesis of a PSMA Inhibitor-Doxorubicin Conjugate for Targeted Prostrate Cancer Therapy," ChemMedChem 2006, 1, pp. 299-302.
JE Dancey. "Therapeutic Targets mTOR and Related Pathways." Cancer Biology & Therapy, 5:9, Sep. 2006, pp. 1065-1073. *
Jeong et al., "Effect of cryoprotectants on the reconstitution of surfactant-free nanoparticles of poly(DL-lactide-co-glycolide)," J. of Microencapsulation. (2005) 22(6):593-601.
JM Koziara, JJ Oh, WS Akers, SP Ferraris, RJ Mumper. "Blood Compatibility of Cetyl Alcohol/Polysorbate-Based Nanoparticles." Pharmaceutical Research, vol. 22 No. 11, Nov. 2005, pp. 1821-1828. *
Kozikowski, Alan P., et al., "Synthesis of Urea-Based Inhibitors as Active Site Probes of Glutamate Carboxypeptidase II: Efficacy as Analgesic Agents," J. Med. Chem. 2004, 47, pp. 1729-1738.
Kozikowski, Alan P., et al., Design of Remarkably Simple, Yet Potent Urea-Based Inhibitors of Glutamate Carboxypeptidase II (NAALADase), J. Med. Chem, 2001, 44, pp. 298-301.
Kwon, "Long Acting Porous Microparticle for Pulmonary Protein Delivery," Int. J. Pharm.(2007) 333:5-9.
Lyseng-Williamson et al. "Docetaxel a Review of its Use in Metastatic Breast Cancer " Drugs. (2005) 65(17):2513-16.
Majer et al., "Synthesis and Biological Evaluation of Thiol-Based Inhibitors of Glutamate Carboxypeptidase II: Discovery of an Orally Active GCP II Inhibitor," J. Med. Chem. (2003) 46:1989-1996.
Maresca, K.P., et al., "A Series of Halogenated Heterodimeric Inhibitors of Prostate Specific Membrane Antigen (PSMA) as Radiolabeled Probes for Targeting Prostate Cancer," J. Med. Chem. (2009) 52(2): 347-57.
Mease, Ronnie C., et al., "N-[N-[(S)-1,3-Dicarboxypropyl] Carbamoyl]-4-[18F]Fluorobenzyl-L-Cysteine, [18F]DCFBC: A New Imaging Probe for Prostate Cancer," Clin. Cancer Res. 2008;14(10) May 15, 2008, pp. 3036-3043.
Merck (Betamethasone, Merck Index (Knovel, copyright 2006, 2012)), 3 pages.
Misra, Preeti, et al., "Production of Multimeric Prostate-Specific Membrance Antigen Small-Molecule Radiotracers Using a Solid-Phase 99m Tc Preloading Strategy," The Journal of Nuclear Medicine, vol. 48, No. 8, pp. 1379-1389., Aug. 2007.
Murugesan et al., "Pegylated Poly(lactide-co-glycolidel (PLGA) Nanoparticulate Delivery of Docetaxel: Synthesis of Diblock Copolymers, Optimization of Preparation Variables on Formulation Characteristics and in Vitro Release Studies." J. Biomed. Nanotechnol. (2007) 3:52-60.
Musumeci et al., "Lyoprotected Nanosphere Formulations for Paclitaxel Controlled Delivery." J. Nanosci. Nanotech. (2006) 6:3118-3125.
Musumeci et al., "PLA/PLGA Nanoparticles for Sustained Release of Docetaxel," Int. J. Pharm. (2006) 325:172-179.
Ojer et al., "Spray-Drying of Poly(anhydride) Nanoparticles for Drug/Antigen Delivery " J. Drug Del. Sci. Tech. (2010) 20(5):353-359.
Oliver, A. Jayne, et al., "Conformational and SAR Analysis of NAALADase and PSMA Inhibitors," Biorganic & Medicinal Chemistry 11 (2003) pp. 4455-4461.
Olivier, "Drug Transport to Brain with Targeted Nanoparticles," The Journal of the American Society for Experimental NeuroTherapeutics, (2005) 2:108-119.
Omelczuk et al., "The Influence of Polymer Glass Transition Temperature and Molecular Weight on Drug Release from Tablets Containing Poly(DL-lactic acid)." Pharm. Res. (1992) 9(1):26-32.
Peracchia, et al., "PEG-coated nanospheres from amphiphilic diblock and multiblock copolymers: Investigation of their drug encapsulation and release characteristics " Journal of Controlled Release (1996) 46:223-231.
Pomper, Martin G., Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, "New Developments in Molecular Imaging of Prostate Cancer," Topical Symposium on: Advanced Molecular Imaging Techniques in the Detection, Diagnosis, Therapy, and Follow-up of Prostate Cancer, Palazzo Barberini, Rome, Dec. 6, 2005.
Pourcelle et al., "PCL-PEG-based Nanoparticles Grafted with GRGDS Peptide: Preparation and Surface Analysis by XPS," Biomacromolecules (2007) 8:3977-3983.
Pulkkinen, M. et al. "Three-Step Tumor Targeting of Paclitaxel Using Biotinylated PLA-PEG Nanoparticles and Avidin-Biotin Technology: Formulation Development and In Vitro Anticancer Activity," Eur. J. Pharm. Biopharm. (2008) 70:66-74.
Riley et al. "Colloidal Stability and Drug Incorporation Aspects of Micellar-Like PLA-PEG Nanoparticles," Colloids Surf. B: Biointer. (1999) 16:147-59.
Sapra, P. et al., "Ligand-Targeted Liposomal Anticancer Drugs," Prog. Lipid Res. (2003) 42:439-462.
Senthilkumar et al., "Long Circulating PEGylated Poly(D,L-lactide-co-glycolide) Nanoparticulate Delivery of Docetaxel to Solid Tumors," J. Drug Target. (2008) 424-435.
Sweetman, "Martindale: The Complete Drug Reference," 33rd ed., 2002, Pharmaceutical Press, entry for Docetaxel, p. 534.
Tamilvanan et al., "Manufacturing Techniques and Excipients Used During the Design of Biodegradable Polymer-based Microspheres Containing Therapeutic Peptide/Protein for Parenteral Controlled Drug Delivery," PDA J. Pharm. Sci. Technol. (2008) 62:125-154.
Tang, Hailun, et al., "Prostate Targeting Ligands Based on N-Acetylated alpha-Linked Acidic Dipeptidase," Biochemical and Biophysical Research Communications 307 (2003), pp. 8-14.
Tang, Hailun, et al., "Prostate Targeting Ligands Based on N-Acetylated α-Linked Acidic Dipeptidase," Biochemical and Biophysical Research Communications 307 (2003), pp. 8-14.
Tobio, M. et al., "Stealth PLA-PEG Nanoparticles as Protein Carriers for Nasal Administration," Pharm. Res. (1998) 15(2):270-275.
Vicari et al., "Paclitaxel Loading in PLGA Nanospheres Affected the in Vitro Drug Cell Accumulation and Antiproliferative Activity," BMC Cancer (2008) 8:212.
XY Jiang, CS Zhou, KW Tang. "Preparation of PLA and PLGA Nanoparticles by Binary Organic Solvent Diffusion Method." J Cent. South Univ. Technol., vol. 10, No. 3, Sep. 2003, pp. 202-206. *
Yamamoto, Y. et al., "Long-Circulating Poly(ethylene glycol)-Poly(D,L-lactide) Block Copolymer Micelles with Modulated Surface Charge," J. Control. Rel. (2001) 77:27-38.
Zhang, Qi et al., "Neointimal Hyperplasia Persists at Six Months After Sirolimus-Eluting Stent Implantation in Diabetic Porcine," Cardiovasc. Diabetol. (2007) 6(16):1-7.
Zhou, Jia, et al., "NAAG Peptidase Inhibitors and Their Potential for Diagnosis and Therapy," Nature Reviews/Drug Discovery, vol. 4, Dec. 2005, pp. 1015-1025.

Cited By (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9579284B2 (en) * 2008-06-16 2017-02-28 Pfizer Inc. Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US20130236500A1 (en) * 2008-06-16 2013-09-12 Bind Biosciences, Inc. Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20140186453A1 (en) * 2008-06-16 2014-07-03 Bind Therapeutics, Inc. Therapeutic Polymeric Nanoparticles with mTOR Inhibitors and Methods of Making and Using Same
US9351933B2 (en) 2008-06-16 2016-05-31 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US9375481B2 (en) 2008-06-16 2016-06-28 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US9393310B2 (en) * 2008-06-16 2016-07-19 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US9579386B2 (en) * 2008-06-16 2017-02-28 Pfizer Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US20120276162A1 (en) * 2008-06-16 2012-11-01 Zale Stephen E Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US8905997B2 (en) 2008-12-12 2014-12-09 Bind Therapeutics, Inc. Therapeutic particles suitable for parenteral administration and methods of making and using same
US20160045608A1 (en) * 2008-12-15 2016-02-18 Bind Therapeutics, Inc. Long circulating nanoparticles for sustained release of therapeutic agents
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US8993549B2 (en) 2011-11-23 2015-03-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8993548B2 (en) 2011-11-23 2015-03-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11793819B2 (en) 2011-11-23 2023-10-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8987237B2 (en) 2011-11-23 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9114145B2 (en) 2011-11-23 2015-08-25 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10675288B2 (en) 2011-11-23 2020-06-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9114146B2 (en) 2011-11-23 2015-08-25 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11103516B2 (en) 2011-11-23 2021-08-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9248136B2 (en) 2011-11-23 2016-02-02 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9006222B2 (en) 2012-06-18 2015-04-14 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11529360B2 (en) 2012-06-18 2022-12-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11110099B2 (en) 2012-06-18 2021-09-07 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9012434B2 (en) 2012-06-18 2015-04-21 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11033626B2 (en) 2012-06-18 2021-06-15 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US11166963B2 (en) 2012-06-18 2021-11-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10639375B2 (en) 2012-06-18 2020-05-05 Therapeuticsmd, Inc. Progesterone formulations
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US8987238B2 (en) 2012-06-18 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11865179B2 (en) 2012-06-18 2024-01-09 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US9877923B2 (en) 2012-09-17 2018-01-30 Pfizer Inc. Process for preparing therapeutic nanoparticles
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US10888516B2 (en) 2012-12-21 2021-01-12 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11351182B2 (en) 2012-12-21 2022-06-07 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11116717B2 (en) 2012-12-21 2021-09-14 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11123283B2 (en) 2012-12-21 2021-09-21 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11622933B2 (en) 2012-12-21 2023-04-11 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11241445B2 (en) 2012-12-21 2022-02-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11497709B2 (en) 2012-12-21 2022-11-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11065197B2 (en) 2012-12-21 2021-07-20 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11304959B2 (en) 2012-12-21 2022-04-19 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10835487B2 (en) 2012-12-21 2020-11-17 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10071100B2 (en) 2014-03-14 2018-09-11 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US9895378B2 (en) 2014-03-14 2018-02-20 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11103513B2 (en) 2014-05-22 2021-08-31 TherapeuticsMD Natural combination hormone replacement formulations and therapies
US10668082B2 (en) 2014-10-22 2020-06-02 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10398708B2 (en) 2014-10-22 2019-09-03 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10912783B2 (en) 2015-07-23 2021-02-09 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US11260027B2 (en) 2015-07-29 2022-03-01 Musc Foundation For Research Development Donor organ pre-treatment formulation
US10266485B2 (en) 2015-09-17 2019-04-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10442756B2 (en) 2015-09-17 2019-10-15 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US11220476B2 (en) 2015-09-17 2022-01-11 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10392341B2 (en) 2015-09-17 2019-08-27 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10799463B2 (en) 2015-12-22 2020-10-13 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10532059B2 (en) 2016-04-01 2020-01-14 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
US11597698B2 (en) 2019-09-19 2023-03-07 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression

Also Published As

Publication number Publication date
WO2010005726A2 (en) 2010-01-14
US20100069426A1 (en) 2010-03-18
WO2010005726A3 (en) 2010-04-22
US8623417B1 (en) 2014-01-07
US20140186453A1 (en) 2014-07-03
US20130344158A1 (en) 2013-12-26
US9579284B2 (en) 2017-02-28

Similar Documents

Publication Publication Date Title
US9579284B2 (en) Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US9351933B2 (en) Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US20160051522A1 (en) Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
US20170035694A1 (en) Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
US9872848B2 (en) Stable formulations for lyophilizing therapeutic particles
KR101706178B1 (en) Drug loaded polymeric nanoparticles and methods of making and using same

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIND BIOSCIENCES, INC.,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZALE, STEPHEN E.;TROIANO, GREG;ALI, MIR MUKKARAM;AND OTHERS;SIGNING DATES FROM 20091120 TO 20091123;REEL/FRAME:023593/0493

Owner name: BIND BIOSCIENCES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZALE, STEPHEN E.;TROIANO, GREG;ALI, MIR MUKKARAM;AND OTHERS;SIGNING DATES FROM 20091120 TO 20091123;REEL/FRAME:023593/0493

AS Assignment

Owner name: BIND THERAPEUTICS, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:BIND BIOSCIENCES, INC.;REEL/FRAME:030573/0152

Effective date: 20130401

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: PFIZER INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BIND THERAPEUTICS, INC.;REEL/FRAME:041261/0721

Effective date: 20160801

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20211224